NPC Natural Product Communications
Transcription
NPC Natural Product Communications
This Issue is Dedicated to Professor Franco F. Vincieri on the Occasion of his 70th Birthday Volume 3. Issue 12. Pages 1941-2166. 2008 ISSN 1934-578X (printed); ISSN 1555-9475 (online) www.naturalproduct.us NPC Natural Product Communications EDITOR-IN-CHIEF DR. PAWAN K AGRAWAL Natural Product Inc. 7963, Anderson Park Lane, Westerville, Ohio 43081, USA agrawal@naturalproduct.us EDITORS PROFESSOR GERALD BLUNDEN The School of Pharmacy & Biomedical Sciences, University of Portsmouth, Portsmouth, PO1 2DT U.K. axuf64@dsl.pipex.com PROFESSOR ALESSANDRA BRACA Dipartimento di Chimica Bioorganicae Biofarmacia, Universita di Pisa, via Bonanno 33, 56126 Pisa, Italy braca@farm.unipi.it PROFESSOR DEAN GUO State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100083, China gda5958@163.com PROFESSOR J. ALBERTO MARCO Departamento de Quimica Organica, Universidade de Valencia, E-46100 Burjassot, Valencia, Spain alberto.marco@uv.es PROFESSOR YOSHIHIRO MIMAKI School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Horinouchi 1432-1, Hachioji, Tokyo 192-0392, Japan mimakiy@ps.toyaku.ac.jp PROFESSOR STEPHEN G. PYNE Department of Chemistry University of Wollongong Wollongong, New South Wales, 2522, Australia spyne@uow.edu.au PROFESSOR MANFRED G. REINECKE Department of Chemistry, Texas Christian University, Forts Worth, TX 76129, USA m.reinecke@tcu.edu PROFESSOR WILLIAM N. SETZER Department of Chemistry The University of Alabama in Huntsville Huntsville, AL 35809, USA wsetzer@chemistry.uah.edu PROFESSOR YASUHIRO TEZUKA Institute of Natural Medicine Institute of Natural Medicine, University of Toyama, 2630-Sugitani, Toyama 930-0194, Japan tezuka@inm.u-toyama.ac.jp ADVISORY BOARD Prof. Viqar Uddin Ahmad Karachi, Pakistan Prof. Øyvind M. Andersen Bergen, Norway Prof. Giovanni Appendino Novara, Italy Prof. Yoshinori Asakawa Tokushima, Japan Prof. Maurizio Bruno Palermo, Italy Prof. Carlos Cerda-Garcia-Rojas Mexico city, Mexico Prof. Josep Coll Barcelona, Spain Prof. Geoffrey Cordell Chicago, IL, USA Prof. Samuel Danishefsky New York, NY, USA Dr. Biswanath Das Hyderabad, India Prof. A.A. Leslie Gunatilaka Tucson, AZ, USA Prof. Stephen Hanessian Montreal, Canada Prof. Michael Heinrich London, UK Prof. Kurt Hostettmann Lausanne, Switzerland Prof. Martin A. Iglesias Arteaga Mexico, D. F, Mexico Prof. Jerzy Jaroszewski Copenhagen, Denmark Prof. Teodoro Kaufman Rosario, Argentina Prof. Norbert De Kimpe Gent, Belgium Prof. Hartmut Laatsch Gottingen, Germany Prof. Marie Lacaille-Dubois Dijon, France Prof. Shoei-Sheng Lee Taipei, Taiwan Prof. Francisco Macias Cadiz, Spain Prof. Anita Marsaioli Campinas, Brazil Prof. Imre Mathe Szeged, Hungary Prof. Joseph Michael Johannesburg, South Africa Prof. Ermino Murano Trieste, Italy Prof. Virinder Parmar Delhi, India Prof. Luc Pieters Antwerp, Belgium Prof. Om Prakash Manhattan, KS, USA Prof. Peter Proksch Düsseldorf, Germany Prof. William Reynolds Toronto, Canada Prof. Raffaele Riccio Salerno, Italy Prof. Ricardo Riguera Santiago de Compostela, Spain Prof. Satyajit Sarker Coleraine, UK Prof. Monique Simmonds Richmond, UK Prof. Valentin Stonik Vladivostok, Russia Prof. Hermann Stuppner Innsbruck, Austria Prof. Apichart Suksamrarn Bangkock, Thailand Prof. Hiromitsu Takayama Chiba, Japan Prof. Karin Valant-Vetschera Vienna, Austria Prof. Peter G. Waterman Lismore, Australia Prof. Paul Wender Stanford, USA INFORMATION FOR AUTHORS Full details of how to submit a manuscript for publication in Natural Product Communications are given in Information for Authors on our Web site http://www.naturalproduct.us. Authors may reproduce/republish portions of their published contribution without seeking permission from NPC, provided that any such republication is accompanied by an acknowledgment (original citation)-Reproduced by permission of Natural Product Communications. Any unauthorized reproduction, transmission or storage may result in either civil or criminal liability. The publication of each of the articles contained herein is protected by copyright. Except as allowed under national “fair use” laws, copying is not permitted by any means or for any purpose, such as for distribution to any third party (whether by sale, loan, gift, or otherwise); as agent (express or implied) of any third party; for purposes of advertising or promotion; or to create collective or derivative works. Such permission requests, or other inquiries, should be addressed to the Natural Product Inc. (NPI). A photocopy license is available from the NPI for institutional subscribers that need to make multiple copies of single articles for internal study or research purposes. To Subscribe: Natural Product Communications is a journal published monthly. 2008 subscription price: US$1,395 (Print, ISSN# 1934-578X); US$1,095 (Web edition, ISSN# 1555-9475); US$1,795 (Print + single site online). Orders should be addressed to Subscription Department, Natural Product Communications, Natural Product Inc., 7963 Anderson Park Lane, Westerville, Ohio 43081, USA. Subscriptions are renewed on an annual basis. Claims for nonreceipt of issues will be honored if made within three months of publication of the issue. All issues are dispatched by airmail throughout the world, excluding the USA and Canada. Natural Product Communications Vol. 3 (12) 2008 Published online (www.naturalproduct.us) Editorial In Honor of the 70th Birthday of Professor Franco Francesco Vincieri It is our great privilege and pleasure to introduce this issue, which is dedicated to Professor Franco Francesco Vincieri, Faculty of Pharmacy, Department of Pharmaceutical Sciences, of the University of Florence, on the occasion of his 70th birthday. His research has been based on numerous aspects of modern pharmacognosy, offering valuable contributions to pharmacognosy (pharmaceutical biology) and analytical phytochemistry, and more recently pharmaceutical technology applied to herbal drug preparations. He is author of more than 180 publications in international scientific journals, several books and chapters of books and a great number of contributions in congress proceedings. Prof. Vincieri’s efforts to promote quality and modernization, including the technological aspects of herbal medicinal products, have been acknowledged by the Italian Government through the financing of projects related to optimization of the technological and biopharmaceutical aspects of Herbal Medicinal Products and Botanical Health Products to improve their quality and safety of use, as well as awards from national and international scientific societies including ESCOP (European Scientific Cooperative on Phytotherapy) and APV (Abeitsgemeinschaft für Pharmazeuitsche Verfahrenstechnik E.V.). This issue, on occasion of his 70th Birthday on October 12th 2008, represents a tribute for his outstanding scientific contributions and an opportunity to express our congratulations and warm wishes from us, as well as his colleagues and friends. Our personal thanks go also to all the authors, mostly of them Franco’s friends, and reviewers who have contributed to the success of this special issue. Anna Rita Bilia Pawan K. Agrawal Editor in chief Natural Product Communications Vol. 3 (12) 2008 Published online (www.naturalproduct.us) Preface Franco Francesco Vincieri: A succesful example of youthful enthusiasm for research and education This issue of Natural Product Communications recognizes Professor Franco Francesco Vincieri on the occasion of his 70th birthday celebrated on October 12, 2008. I have invited some dear colleagues to present him in writing. First of all, the Director of the Department of Pharmaceutical Sciences, Prof. Massimo Bambagiotti Alberti, and the Dean of the Faculty of Pharmacy, Prof. Sergio Pinzauti, who have also been good friends since the beginning of his academic career. Prof. Vincieri also served for many years in the Italian Society of Phytochemistry (SIF) and the European Scientific Committee (ESCOP) and thus I asked the SIF president, Prof. Cosimo Pizza, and the Chair of ESCOP, Dr. Barbara Steinhoff, to comment on his role in these organizations. Prof. Anna Rita Bilia University of Florence, Department of Pharmaceutical Sciences Via Ugo Schiff 6, 50019 Sesto Fiorentino (FI), Italy I met Franco F. Vincieri for the first time in 1964 when I entered the Institute of Pharmaceutical and Toxicological Chemistry, University of Florence, nowadays called the Department of Pharmaceutical Sciences. Being young and avid researchers, we soon became interested in modern analytical chemistry. As a matter of fact, in the following few years we introduced Gas Chromatography into our Institute with the first instrument acquired by the University of Florence. The analysis of terpenoids and related volatile compounds was the starting point for our ever increasing interest in separation science. Gas Chromatography-Mass Spectrometry (GC-MS) was the outcome of the longing to improve our scientific tooling. Again, the acquired instrument was the first GC-MS in the University of Florence. Our research capabilities were boosted accordingly and a new generation of mass spectrometrists was trained and formed in our lab. After 1990 we entered quite different careers, but as we have remained in the same Department, we have proceeded with our collaboration and solid friendship. During that period Franco became increasingly interested in quality aspects of herbal medicinal products and was first involved in the Italian Pharmacopoeia as a member of the Permanent Commission (since 1993) where, since January 2004, he has been President of the Working Group “Herbal Drugs”. He is an expert member of “Consiglio Superiore di Sanità” for the three-year period 2006-2009. Prof. Vincieri was the Italian Delegate, first at HMPWG (Herbal Medicinal Products Working Group) and then at HMPWP (Herbal Medicinal Products Working Party) from 1997 to 2004 at the EMEA. He was assessor of the “Certification procedure for Chemical quality and microbiological purity evaluation” of EDQM (European Directorate for the Quality of Medicines) from 1998 to 2004 and assessor of the “Certification procedure for Herbal Drugs and Herbal Drug Preparations evaluation” of EDQM since 2004. Beginning this year (2008), he is a member of the European Pharmacopoeia Commission of the Traditional Chinese Medicine working Party. Prof. Massimo Bambagiotti Head of Department University of Florence, Department of Pharmaceutical Sciences Via Ugo Schiff 6, 50019 Sesto Fiorentino (FI), Italy Natural Product Communications Vol. 3 (12) 2008 Published online (www.naturalproduct.us) The first time I met Franco F. Vincieri was in June 1967 when I entered as a CNR fellow in the Institute of Pharmaceutical and Toxicological Chemistry. During that time he was involved in the elementary analysis of various products synthetised in the Institute. A strong friendship was quickly created between us and we discovered sharing, apart from our great enthusiasm for synthesis and analysis of new drugs, a passion for photography which led to experimentation of numerous photographical processes of developing and printing. It was a “chemical” way to cultivate through images, during our free time, our aesthetics, which I recall as distinguishing. In the Seventies he passed from being professor in charge of the "Laboratory of Extractive and Synthetic Drug Preparation" to being an associate, then supernumerary professor of "Phytopharmacy". Now he is full professor of Pharmaceutical Technology, Socio-economics and Legislation. Over the last 45 years his teaching skills have been very much appreciated by generations of students. He is quite active in the University of Florence, taking on various institutional roles such as Vice Dean of the Faculty of Pharmacy since 2001, Director of the graduate school of “Herbal Technology” from 2000 to 2006, Director of the Professional Specialization Course “Expert Technician in Officinal Herbs” (collaboration between the University of Florence and the Region of Tuscany) from 2005 to 2006, Director of the post-graduate course “Herbal Medicinal Products of Traditional Chinese Medicine” since 2005 and Director of the post-graduate school of “Hospital Pharmacy ” from 1994 to 2007. His research has been based on numerous aspects and he is author or coauthor of more than 180 publications in international scientific journals, several books and chapters of books, and a great number of contributions in congress proceedings. Prof. Sergio Pinzauti Editor, Journal of Pharmaceutical and Biomedical Analysis Dean, Faculty of Pharmacy University of Florence, Department of Pharmaceutical Sciences Via Ugo Schiff 6, 50019 Sesto Fiorentino (FI), Italy It is an honour and a pleasure for me, as Chairman of the Italian Society of Phytochemistry (SIF) and also as a colleague and friend, to send wishes to Prof. Franco Vincieri, also on behalf of the Society, on the occasion of his 70th birthday. Prof. Vincieri, at the beginning of 80’s founded the Italian Society of Phytochemistry together with other distinguished scientists in the field and he was a member of the board for many years. The Italian Society of Phytochemistry has the aim of promoting knowledge of the plant kingdom by phytochemical, pharmacological, toxicological, food, industrial and ecological point of view and the cooperation with other national and international organizations pursuing the same objectives. Prof. Franco Vincieri has always participated in all the SIF activities, contributing with his considerable scientific profile and his ability as organizer to various national and international events, paying particular attention to the initiatives devoted to young scientists. In the period 1998-2000 he was chairman of our Society, promoting a number of successful scientific activities and workshops, for example on Ginkgo biloba, on Analytical Methods in Phytochemistry and on Cell Cultures, which attracted a large number of new members to SIF. To Franco, a very outstanding scientist, sincere thanks for all he has done and will continue to do in the future in the field of phytochemistry. Prof. Cosimo Pizza Chairman of Italian Society of Phytochemistry University of Salerno, Department of Pharmaceutical Sciences via Ponte Don Melillo 84084 Fisciano (SA), Italy Natural Product Communications Vol. 3 (12) 2008 Published online (www.naturalproduct.us) For many years, Professor Franco F. Vincieri has been very active in ESCOP, the European Scientific Cooperative on Phytotherapy. Starting already in 1991 at an early stage of this scientific organisation, he represented the Società Italiana di Fitochimica as host of the 2nd ESCOP Symposium which took place in Milan in March 1992 and chaired the meeting of the ESCOP Council. The colleagues from the Scientific Committee remember well the fruitful ESCOP meetings consisting of detailed work and long discussions on draft monographs which are intended to provide harmonised assessment criteria for efficacy and safety of herbal medicinal products in Europe, but also beautiful visits to botanical gardens and other places of interest. They gratefully accepted his kind invitation to the Florence meeting in May 1996 which took place in the Scuolà di Sanità Militare, the concert of "I Mandolinisti Fiorentini" in the evening being one of the special highlights of this weekend. When Franco Vincieri handed over his ESCOP seat to Professor Anna Rita Bilia in 1997, ESCOP offered him to continue his activities as a member of the Board of Supervising Editors which he kindly accepted. Since 2004 he is also member of the ESCOP Research Committee. In this position he reviews all the monographs produced by the Scientific Committee and comments on each of them before publication. Due to his long-term experience in pharmacognosy and related scientific areas his suggestions for modifications are highly appreciated by the Committee. The members of the ESCOP Scientific Committee would like to congratulate Franco Vincieri on his 70th birthday and express their sincere thanks for all his contributions to the work of ESCOP. They are looking forward to further enjoyable collaboration in the future and would like to thank him for being such a good colleague. Dr. Barbara Steinhoff Bundesverband der Arzneimittel-Hersteller e.V. Postfach 20 12 55 53142 Bonn, Germany Happy birthday from all of us, colleagues and friends! Natural Product Communications 2008 Volume 3, Number 12 Contents Page 1968-2008: 40 Years of Franco F. Vincieri’s Natural Products Research Anna Rita Bilia 1941 Effects of Terpenoids from Salvia willeana in Delayed-type Hypersensitivity, Human Lymphocyte Proliferation and Cytokine Production Anna Vonaparti, Anastasia Karioti, María C. Recio, Salvador Máñez, José L. Ríos, Eleani Skaltsa and Rosa M. Giner 1953 Characterization of By-products of Saffron (Crocus sativus L.) Production Pamela Vignolini, Daniela Heimler, Patrizia Pinelli, Francesca Ieri, Arturo Sciullo and Annalisa Romani 1959 Antitrypanosomal and Antileishmanial Activities of Organic and Aqueous Extracts of Artemisia annua Anna Rita Bilia, Marcel Kaiser, Franco Francesco Vincieri and Deniz Tasdemir 1963 Secondary Metabolites from the Roots of Salvia palaestina Bentham Antonio Vassallo, Ammar Bader, Alessandra Braca, Angela Bisio, Luca Rastrelli, Francesco De Simone and Nunziatina De Tommasi 1967 Cancer Chemopreventive Potential of Humulones and Isohumulones (Hops α- and Iso-α-acids): Induction of NAD(P)H:Quinone Reductase as a Novel Mechanism Gregor Bohr, Karin Klimo, Josef Zapp, Hans Becker and Clarissa Gerhäuser 1971 A Polar Cannabinoid from Cannabis sativa var. Carma Giovanni Appendino, Anna Giana, Simon Gibbons, Massimo Maffei, Giorgio Gnavi, Gianpaolo Grassi and Olov Sterner 1977 HPLC-DAD-MS Fingerprint of Andrographis paniculata (Burn. f.) Nees (Acanthaceae) Sabrina Arpini, Nicola Fuzzati, Andrea Giori, Emanuela Martino, Giacomo Mombelli, Luca Pagni and Giuseppe Ramaschi 1981 Diterpenoid Alkaloids and Phenol Glycosides from Aconitum naviculare (Brühl) Stapf. Stefano Dall’Acqua, Bharat B. Shrestha, Mohan Bikram Gewali, Pramod Kumar Jha , Maria Carrara and Gabbriella Innocenti 1985 Inhibition of PGHS-1 and PGHS-2 by Triterpenoid Acids from Chaenomelis fructus Eveline Reininger and Rudolf Bauer 1991 Preparative Isolation of Antimycobacterial Shoreic Acid from Cabralea canjerana by High Speed Countercurrent Chromatography Gilda G. Leitão, Lisandra F. Abreu, Fernanda N. Costa, Thiago B. Brum, Daniela Fernandes Ramos, Pedro Eduardo A. Silva, Maria Cristina S. Lourenço and Suzana G. Leitão 1995 Antiplasmodial Effects of a few Selected Natural Flavonoids and their Modulation of Artemisinin Activity Anna Rita Bilia, Anna Rosa Sannella, Franco Francesco Vincieri, Luigi Messori, Angela Casini, Chiara Gabbiani , Carlo Severini and Giancarlo Majori 1999 Comparative Analysis of Antimalarial Principles in Artemisia annua L. Herbal Drugs from East Africa Silvia Lapenna, Maria Camilla Bergonzi, Franco Francesco Vincieri and Anna Rita Bilia 2003 Continued Overleaf Natural Product Communications Vol. 3 (12) 2008 Published online (www.naturalproduct.us) In vitro Apoptotic Bioactivity of Flavonoids from Astragalus verrucosus Moris Joseph A. Buhagiar, Alessandra Bertoli, Marie Therese Camilleri-Podesta and Luisa Pistelli 2007 Qualitative Profile and Quantitative Determination of Flavonoids from Crocus sativus L. Petals by LC-MS/MS Paola Montoro, Carlo I. G. Tuberoso, Mariateresa Maldini, Paolo Cabras and Cosimo Pizza 2013 HPLC/DAD/ESI-MS Analysis of Non-volatile Constituents of Three Brazilian Chemotypes of Lippia alba (Mill.) N. E. Brown Patrícia Timóteo, Anastasia Karioti, Suzana G. Leitão, Franco Francesco Vincieri and Anna Rita Bilia 2017 Optimization and Validation of an HPLC–Method for Quality Control of Pueraria lobata Root Lidiya Bebrevska, Mart Theunis, Arnold Vlietinck, Luc Pieters and Sandra Apers 2021 Pharmacokinetics of Luteolin and Metabolites in Rats Sasiporn Sarawek, Hartmut Derendorf and Veronika Butterweck 2029 Complete Characterization of Extracts of Onopordum illyricum L. (Asteraceae) by HPLC/PDA/ESIMS and NMR Luisella Verotta, Laura Belvisi, Vittorio Bertacche and Maria Cecilia Loi 2037 Phenolic Profiles of Four Processed Tropical Green Leafy Vegetables Commonly Used as Food Sule Ola Salawu, Marzia Innocenti, Catia Giaccherini, Afolabi Akintunde Akindahunsi and Nadia Mulinacci 2043 (Bio)Sensor Approach in the Evaluation of Polyphenols in Vegetal Matrices M. Camilla Bergonzi, Maria Minunni and Anna Rita Bilia 2049 In vitro Radical Scavenging and Anti-Yeast Activity of Extracts from Leaves of Aloe Species Growing in Congo Annalisa Romani, Pamela Vignolini, Laura Isolani, Sara Tombelli, Daniela Heimler, Benedetta Turchetti and Pietro Buzzini 2061 Antioxidant Principles and Volatile Constituents from the North-western Iberian mint “erva-peixeira”, Mentha cervina Matteo Politi, César L Rodrigues, Maria S Gião, Manuela E Pintado and Paula ML Castro 2065 Chemical Composition of Thymus serrulatus Hochst. ex Benth. Essential Oils from Ethiopia: a Statistical Approach Bruno Tirillini, Roberto Maria Pellegrino, Mario Chessa and Giorgio Pintore 2069 GC MS Analysis of the Volatile Constituents of Essential Oil and Aromatic Waters of Artemisia annua L. at Different Developmental Stages Anna Rita Bilia, Guido Flamini, Fabrizio Morgenni, Benedetta Isacchi and Franco FrancescoVincieri 2075 Do Non-Aromatic Labiatae Produce Essential Oil? The Case Study of Prasium majus L. Claudia Giuliani, Roberto Maria Pellegrino, Bruno Tirillini and Laura Maleci Bini 2079 Olive-oil Phenolics and Health: Potential Biological Properties Francesco Visioli, Francesca Ieri, Nadia Mulinacci, Franco F. Vincieri and Annalisa Romani 2085 Traceability of Secondary Metabolites in Eucalyptus and Fagus Wood derived Pulp and Fiber Aline Lamien-Meda, Karin Zitterl-Eglseer, Heidrun Fuchs and Chlodwig Franz 2089 Potential Anticancer Activity Against Human Epithelial Cancer Cells of Peumus boldus Leaf Extract Juan Garbarino, Nicolas Troncoso, Giuseppina Frasca, Venera Cardile and Alessandra Russo 2095 Antihyperalgesic Effect of Eschscholzia californica in Rat Models of Neuropathic Pain Elisa Vivoli, Anna Maidecchi, Anna Rita Bilia, Nicoletta Galeotti, Monica Norcini and Carla Ghelardini 2099 Natural Product Communications Vol. 3 (12) 2008 Published online (www.naturalproduct.us) Problems in Evaluating Herbal Medicinal Products Jozef Corthout 2103 Impurities in Herbal Substances, Herbal Preparations and Herbal Medicinal Products, IV. Heavy (toxic) Metals SFSTP Commission, Didier Guédon, Michèle Brum, Jean-Marc Seigneuret, Danièle Bizet, Serge Bizot, Edmond Bourny, Pierre-Albert Compagnon, Hélène Kergosien, Luis Georges Quintelas, Jerôme Respaud, Olivier Saperas, Khalil Taoubi and Pascale Urizzi 2107 Review/Account A Fresh Insight into the Interaction of Natural Products with Pregnane X Receptor Salvador Máñez 2123 Natural Products as Gastroprotective and Antiulcer Agents: Recent Developments Rosa Tundis, Monica R Loizzo, Marco Bonesi, Federica Menichini, FilomenaConforti, Giancarlo Statti and Francesco Menichini 2129 Phytochemistry and Pharmacology of Boronia pinnata Sm. MassimoCurini, Salvatore Genovese, Luigi Menghini, Maria Carla Marcotullio and Francesco Epifano 2145 Therapeutic Potential of Kalanchoe Species: Flavonoids and other Secondary Metabolites Sônia S. Costa, Michelle F. Muzitano, Luiza M. M. Camargo and Marcela A. S. Coutinho 2151 Manuscripts in Press 2165 Natural Product Communications Vol. 3 (12) 2008 Published online (www.naturalproduct.us) LIST OF AUTHORS Abreu, LF ................. 1995 Acqua, SD ................ 1985 Akindahunsi, AA...... 2043 Apers, S .................... 2021 Appendino, G ........... 1977 Arpini, S ................... 1981 Bader, A ................... 1967 Bauer, R.................... 1991 Bebrevska, L............. 2021 Becker, H.................. 1971 Belvisi, L .................. 2037 Bergonzi, MC .. 2003,2049 Bertacche, V ............. 2037 Bertoli, A .................. 2007 Bilia, AR. 1941,1963,1999 2003,2017,2049,2075,2099 Bini, LM ................... 2079 Bisio, A..................... 1967 Bizet, D..................... 2107 Bizot, S ..................... 2107 Bohr, G ..................... 1971 Bonesi, M ................. 2129 Bourny, E ................. 2107 Braca, A.................... 1967 Brum, M ................... 2107 Brum, TB.................. 1995 Buhagiar, JA............. 2007 Butterweck, V .......... 2029 Buzzini, P ................. 2061 Cabras, P................... 2013 Camargo, LMM........ 2151 Cardile, V ................. 2095 Carrara, M ................ 1985 Casini, A................... 1999 Castro, PML ............. 2065 Chessa, M ................. 2069 Commission, SFSTP 2107 Compagnon, P .......... 2107 Conforti, F ................ 2129 Corthout, J ................ 2103 Costa, FN.................. 1995 Costa, SS .................. 2151 Coutinho, MAS 2151 Curini, M .................. 2145 Derendorf, H .............2029 Epifano, F..................2145 Flamini, G .................2075 Franz, C .....................2089 Frasca, G ...................2095 Fuchs, H ....................2089 Fuzzati, N ..................1981 Gabbiani , C...............1999 Galeotti, N ................2099 Garbarino, J ...............2095 Genovese, S...............2145 Gerhäuser, C..............1971 Gewali, MB ...............1985 Ghelardini, C .............2099 Giaccherini, C ...........2043 Giana, A ....................1977 Gião, MS ...................2065 Gibbons, S .................1977 Giner, RM .................1953 Giori, A......................1981 Giuliani, C .................2079 Gnavi, G ....................1977 Grassi, G....................1977 Guédon, D .................2107 Heimler, D.................1959 Heimler, D.................2061 Ieri, F ............... 1959,2085 Innocenti, G...............1985 Innocenti, M ..............2043 Isacchi, B...................2075 Isolani, L ...................2061 Leitão, SG1995,2003,2017 Loi, MC .....................2037 Loizzo, MR ...............2129 Lourenço, MCS.........1995 Maffei, M ..................1977 Maidecchi, A.............2099 Majori, G ...................1999 Maldini, M ................2013 Máñez, S....................1953 Máñez, S....................2123 Marcotullio, MC .......2145 Martino, E .................1981 Menghini, L...............2145 Menichini, F ..............2129 Messori, L .................1999 Minunni, M ...............2049 Mombelli, G ..............1981 Montoro, P ................2013 Morgenni, F...............2075 Mulinacci, N.....2043,2085 Muzitano, MF............2151 Norcini, M ................2099 Pagni, L .....................1981 Pellegrino, RM .2069,2079 Pieters, L ...................2021 Pinelli, P ....................1959 Pintado, ME ..............2065 Pintore, G ..................2069 Pistelli, L ...................2007 Pizza, C .....................2013 Podesta, MTC............2007 Politi, M.....................2065 Quintelas, LG ............2107 Jha, PK ......................1985 Kaiser, M...................1963 Karioti, A......... 1953,2017 Kergosien, H .............2107 Klimo, K....................1971 Lamien-Meda, A .......2089 Lapenna, S.................2003 Ramaschi, G ..............1981 Ramos, DF ................1995 Rastrelli, L.................1967 Recio, MC .................1953 Reininger, E ..............1991 Respaud, J .................2107 Ríos, JL .....................1953 Rodrigues, CL ...........2065 Romani, A1959,2061,2085 Russo, A ....................2095 Salawu, SO................2043 Sannella, AR .............1999 Saperas, O .................2107 Sarawek, S.................2029 Sciullo, A ..................1959 Seigneuret, J ..............2107 Severini , C ................1999 Shrestha, BB .............1985 Silva, PE....................1995 Simone, FD ...............1967 Skaltsa, E...................1953 Statti, G .....................2129 Sterner, O ..................1977 Taoubi, K ..................2107 Tasdemir, D...............1963 Theunis, M ................2021 Timóteo, P.................2017 Tirillini, B.........2069,2079 Tombelli, S................2061 Tommasi, ND............1967 Troncoso, N...............2095 Tuberoso, CIG...........2013 Tundis, R...................2129 Turchetti, B ...............2061 Urizzi, P ....................2107 Vassallo, A ................1967 Verotta, L ..................2037 Vignolini, P ......1959,2061 Vincieri, FF .....1963,1999, 2003 ,2017,2075,2085 Visioli, F....................2085 Vivoli, E ....................2099 Vlietinck, A...............2021 Vonaparti, AV...........1953 Zapp, J.......................1971 Zitterl-Eglseer, K ......2089 NPC Natural Product Communications 1968-2008: 40 Years of Franco F. Vincieri’s Natural Products Research 2008 Vol. 3 No. 12 1941 - 1952 Anna Rita Bilia Department of Pharmaceutical Sciences, University of Florence, via Ugo Schiff, 8-50019. Sesto Fiorentino, Florence, Italy ar.bilia@unifi.it Received: November 6th, 2008; Accepted: November 14th, 2008 This paper presents an overview of Prof. Vincieri’s accomplishments in his career as a researcher in the field of pharmacognosy (pharmaceutical biology), analytical phytochemistry and pharmaceutical technology applied to herbal drug preparations at the Department of Pharmaceutical Sciences of the University of Florence. This article is a recognition of his valuable contributions to these research fields, especially for his outstanding and innovative interdisciplinary studies on the quality control of herbal drugs, herbal drug preparations, herbal medicinal products, botanical food supplements, and some “special foods” such as grapes, wines, olives and olive oil. Keywords: Franco Francesco Vincieri, Department of Pharmaceutical Sciences, University of Florence, pharmacognosy (pharmaceutical biology), analytical phytochemistry and pharmaceutical technology, herbal drug preparations. This issue of Natural Product Communications is dedicated to the 70th birthday of Franco Francesco Vincieri, Full Professor at the University of Florence, Department of Pharmaceutical Sciences. It has been my special honor to prepare this paper concerning some of his outstanding achievements in the field of pharmacognosy (pharmaceutical biology), analytical phytochemistry and pharmaceutical technology applied to herbal drug preparations, especially for his important and innovative interdisciplinary studies on the quality control of herbal drugs, herbal drug preparations, herbal medicinal products, botanical food supplements and some “special foods” such as grapes, wines, olives and olive oil. I am grateful for the comments and contributions of all colleagues, former and present students, and friends who participated in this special issue for their comments and contributions, and in particular for the construction and revision of this article. My special thanks to Anastasia Karioti for her patient collaboration in reading and commenting on some parts of the paper. Scientifically Prof. Vincieri is a very polyhedral and curious scientist, “a sort of active vulcan”, ready to start a new experience, even if it is an expedition to the African desert (Figure 1). Most of his success has come from his own intense efforts and his extreme Figure 1: Franco duing an expedition through Sahara from Algeria to Giordania. The Sheik, Jeleil al-Deisah, with one of his wives, Hadra, and his childrem is recognising some herbal drugs collected by Franco (in the middle) and his assistent Giacalone (on the right). versatility and passion for research, which led to his many accomplishments, awards and memberships which highlight his extraordinary abilities, not only as a scientist, but also as a person. For these reasons, in 1997, when as a post-doc I moved from the University of Pisa for a permanent position as Researcher at the Department of Pharmaceutical Sciences in Florence, he has had a deep personal and professional impact on my life, a sentiment that is shared by all his former and present students, some of whom are now either post-docs or colleagues in this Department. 1942 Natural Product Communications Vol. 3 (12) 2008 Bilia Figure 3: Sedum telephium L. ssp. maximum Schinz & Thell. Figure 2: ESCOP meeting at Hailsham (UK) in June 1995. Franco talking to the late Prof. Hein Zeylstra (on the left of the picture) and Prof. Finn Sandberg. Among his publications, including several chapters and books (most of them used as text books for undergraduate, postgraduate, master’s and PhD students), I have selected papers concerning several projects which represent the scope and breadth of his work. In addition, these publications are considered milestones for the development of many scientific activities, not only of his research group but also of the Department of Pharmaceutical Sciences, which attracted many young researchers from all parts of the world, and led to finance for his work from private and public pharmaceutical companies. His firm belief that both basic and applied research are equally important is reflected in his efforts in 1981 when he was instrumental in organizing the Italian Society of Phytochemistry (Società Italiana di Fitochimica, SIF), an association of both university and company researchers. He was for many years the Italian delegate of the EMEA (the European Medicines Agency) and ESCOP (the European Scientific Cooperative on Phytotherapy) Scientific Committee working to assess criteria for efficacy and safety of herbal medicinal products in Europe. At the beginning of his academic career (in the late Sixties-early Seventies) he worked with his colleague and friend, Prof. Massimo Bambagiotti Alberti (the current Director of the Department), on the analysis of terpenoids and related volatile compounds which marked the starting point for their ever-increasing interest in separation science. After a few years, Prof. Silvia Coran joined them, and later on Prof. Gloriano Moneti and Prof. Valerio Giannellini. In a short timethe group became well known at the University of Florence for their modern analytical chemistry, thanks to the introduction of the first GC- MS at the University of Florence. There are several papers [1-7] related to volatiles from Pinus species using diverse spectroscopic approaches for the structural elucidation of the components In the same period his friendship and collaboration [8,9] with Prof. Sergio Pinzauti, today the Dean of the Faculty of Pharmacy, also began. Prof. Vincieri’s instrumental ability attracted research groups of the University of Florence interested in diverse disciplines, and in particular Prof. Maria Teresa Vincenzini, one of the most outstanding scientists in Florence, interested in the effects of some natural constituents on the biochemistry of enzymatic systems (dehydrogenases, lyases), especially on germination of several medicinal plants. Their collaboration was confirmed by many papers published during the Sevetnties [10-16]. The studies of Sedum telephium L. ssp. maximum Schinz & Thell. and Oenanthe aquatica (L.) Poiret marked the beginning of analysis of natural constituents from the plant kingdom for the Department, in addition to the analysis of pharmaceutical products. S. telephium (Crassulaceae) (Figure 3) is largely diffused in traditional medicine, especially in Tuscany, as a remedy for the local treatment of wounds and inflammatory diseases of the skin [17]. The term “telephium” is probably related to its vulnerary properties, introduced by Plinius, who first reported this plant as the herbal drug used to cure the leg wounds of King Telephium. The leaves (without the external cuticle, Figure 4) are reported in “The medicatis herbarum facultatibus” by Fulgenzo Vitman (1770), a monk of Vallombrosa (Florence), with this description: "Ulcera detergit...(clean ulcers)...et ad cicatricem perducit ... ( and support Franco F. Vincieri’s natural product research Natural Product Communications Vol. 3 (12) 2008 1943 two new ones, kaempferol-3-O-β-neohesperidoside7-O-α-rhamnoside and quercetin-3-O-βneohesperidoside-7-O-α-rhamnoside, which could contribute to the anti-inflammatory activity of Sedum [21]. Figure 4: Preparation of Sedum leave before its application. cicatrization) ..tumorum suppurationem promovet ... (favour the suppurative phlogistic process) .. et dolores mitigat (and sooth pain)”. At the beginning of the Eighties, the properties of either the fresh or deep-frozen leaves, with the peel of the inferior part removed, were rediscovered and confirmed by a clinician, Dr Sergio Balatri, first at the Emergency Unit of the Torte Galli Hospital and later at San Giovanni di Dio Hospital, Florence in the treatment of various local inflammatory conditions, including whitlow, abscesses, complicated wounds, burns, poor cicatrisation, cysts, ulcerous phlebitis, and horniness (Figure 5) [18]. Figure 5: Application of the fresh peeled leaves of Sedum telephium to a withlow resulting in rapid healing, shown in "before" and "after" pictures. Dr Balatri put forward to Prof. Vincieri the need for chemical analysis of the plant, the testing of its extracts to recognise the compounds responsible for the activity, and the preparation of modern formulations for use in the clinic. Studies started with the degree thesis investigations of Nadia Mulinacci [19], currently associate professor in the Vincieri’s team, and collaboration with Prof. Hildeberg Wagner (University of Munich, Germany). This led to the isolation and identification of polysaccharides from the leaf tissue which had anti-inflammatory potential, including an anticomplementary effect in vitro, induction of TNF-alpha-production, increasing phagocytosis in vitro and in vivo [20]. Two years later the flavonoids were also identified, including Negative-ion fast-atom bombardment mass spectrometry was employed in the identification of flavonol glycosides directly in the juice [22]. Other methodologies were proposed for the qualitative and quantitative determination of flavonol glycosides, in particular a study in collaboration with Prof. Hermann Stuppner and coworkers (University of Innsbruck, Austria), which used MEKC analysis which was compared with HPLC-MS using electrospray ionization (ESI) interface [23]. In another collaborative study with Prof. Francesco Bonina (University of Catania, Italy), in-vitro and invivo studies suggested that, both the total lyophilized juice and, in particular, the lyophilized flavonoidic fraction, but not the lyophilized polysaccharidic fraction of the leaves, have photoprotective effects against UVB-induced skin damage [24]. Further studies with Prof. Renato Pirisino, Prof. Laura Raimondi and Prof. Maria Grazia Banchelli of the Department of Preclinical and Clinical Pharmacology of the University of Florence revealed that total Sedum juice strongly inhibited cell adhesion to laminin and fibronectin (EC50 1.03±0.12 mg mL-1). This anti-adhesive feature was concentrated mainly in the two polysaccharide fractions (EC50 values between 0.09 and 0.44 mg mL-1). The flavonol fractions did not seem to contribute to this effect [25]. According to the phytochemical, biological and pharmacological findings, some simple preparations were developed using either the fresh or lyophilized juice, and fractions. However, in contrast to the fact that extracts can represent the best way to select constituents of plants to be used for medicinal purposes, in the case of Sedum it was not possible to obtain the active fraction or constituents and prepare a valid formulation from them. The leaves still represent the best form for application, as a natural plaster. All efforts to update this simple formulation have been fruitless. Another interesting medicinal plant investigated by Prof. Vincieri is O. aquatica (Apiaceae, Figure 6). Fruits, their alcoholic extract and essential oil are widely reported as a valuable remedy for dyspepsia, for the treatment of chronic pectoral affections as an 1944 Natural Product Communications Vol. 3 (12) 2008 Figure 6: Oenanthe aquatica (L.) Poiret. expectorant, for intermittent fevers, as a diuretic and for obstinate ulcers [26]. The infusion of the fruits was also reported by the “Farmacopea italiana del Regno” until the 4th Edition (1920). However, it has been known for a long time that the fruits can cause vertigo, dizziness, inebriation, dull pains in the head and other narcotic effects, as reported in “A Further Account of the Poisonous Effects of Oenanthe aquatica Succo Viroso Crocante of Lobel, or Hemlock Dropwort” [27]. Problems related to this herbal drug and preparations were diverse, all related to its safe use. Initial studies by Prof. Vincieri provided definitive information on the composition of the essential oil and the light petroleum (40-60°) extract of the fruit by a combination of different techniques: GLC, UV and IR spectroscopy and MS [28]. A group of C15 hydrocarbon and oxygenated polyacetylenes, including three new compounds, were also isolated and structurally identified on the basis of spectroscopic evidence. Due to their extreme sensitivity to air, heat and light, removal of the solvents and spectroscopic manipulation of the sample was performed by a home-made device to directly measure NMR spectra, similar to the modern concept of the hyphenated systems. IR spectra were performed in the crystalline state at liquid nitrogen temperature to observe the out-of-plane vibration band of the cis-double bond which, in the liquid phase, forms one broad band with the -CH2-rocking vibration [29,30]. With the aim of finding a rapid method of characterization of polyacetylenes, second-derivative UV spectra of polyacetylenes were studied with Prof. Mario Pio Marzocchi and Prof. Giulietta Smulevich, colleagues in the Department of Chemistry of the University of Florence. This method was useful for the complete structural identification Bilia of their chromophores, including the stereoisomerism of the double bonds [31]. The second-derivative technique was also applied to a series of related naturally-occurring polyenynes, whose chromophoric fragments ranged from three to six conjugated groups. These results account for the structural characteristics, including steroisomerism, of a given sequence of triple and double bonds providing a complete fingerprint of the polyenyne chromophore [32]. In another paper, the application of the MS-MS technique for the rapid monitoring of some γ-butyrolactone ring and related lignans of O. aquatica fruits infusion was reported [33]. Finally, the GC-MS technique led to the identification of the constituents of the fruit tincture.Ten polyacetylenes, three lignans (derivatives of matairesinol), numerous monoterpenes, including phellandrene and cryptone, dillapiole and small amounts of sterols were identified [34]. Due to the low stability of the constituents of the tincture, especially polyacetylenes, a novel system of stabilization of the active principles of tinctures by means of cyclodextrins (α, β, γ-CyD) was also proposed. Microinclusion of the components was found to be incomplete with all three cyclodextrins, however, β-CyD was the most efficient and the stabilization of the most unstable microincluded active principles was verified by means of artificial ageing studies. These studies were possible thanks to an expert pharmaceutical technologist, Prof. Giovanni Mazzi, who joined the Vincieri group during that period. Further studies through artificial membranes also provided evidence for an increase in the permeability of the constituents. These studies on O. aquatica represent a good example of an interdisciplinary approach, including the enhanced biopharmaceutical properties of the formulated phytocomplex, representing a true milestone in this field of research [35,36]. During the 1980s-1990s, Vincieri expanded his chemistry program to cover many other herbal products and in particular vegetal matrices of interest in the biological and/or alimentary fields aiming at the development of specific methods of extraction, fractionation, isolation and characterization of potentially interesting secondary metabolites for the pharmaceutical, alimentary and cosmetic fields. Two typical Tuscan species with agro-alimentary interest, Vitis vinifera L. (leaves, fruits, wines) and Olea europaea L. (leaves, olives, oil, olive residues and waste waters), have been included in his investigations. Other plants belonging to the Mediterranean maquis, such as Myrtus communis L., Franco F. Vincieri’s natural product research Natural Product Communications Vol. 3 (12) 2008 1945 Figure 7: Sangiovese, a typical grape variety of Chanti DOCG wine. Pistacia lentiscus L., Phillyrea latifolia L., Ligustrum vulgare L., Ligustrum sinensis L., Fraxinum ornus L., Arbutus unedo L. were also studied [37-39]. Studies of polyphenols in wines and grapes were developed by Dr Alessandro Baldi and Prof. Annalisa Romani, former PhD students, and Romani later as a post-doc, researcher and associate professor. The key studies are represented by the development of an analytical method for anthocyanins of Vitis vinifera L. (Vitaceae) [40,41].The pool of anthocyanins contained in the berry skins of different cultivars of V. vinifera was taken as a research model to investigate the possible application of HPLC/MS to anthocyanins. The interface chosen was the API (atmospheric pressure ionization) ion spray interface coupled with a quadrupole mass spectrometer, which allows ambient pressure ionization and the use of any aqueous eluent.The use of this technique made it possible to obtain the mass spectra of all the anthocyanin compounds present in the extracts under investigation, even those occurring in traces or some coeluted ones. Studies were first carried out on certificated clones belonged to two varieties commonly used for the production of the Chianti DOCG red wine: Sangiovese (clone SS-F9-A5-48, [40] and Colorino (clone Nipozzano 6) [41]. The developed HPLC method led to the identification of the 3-glucosides, the 3-acetylglucosides, and the 3p-coumaroylglucosides of delphinidin, cyanidin, petunidin, peonidin, and malvidin, already known in the literature. Two 3-caffeoylglucoside derivatives were identified too, and for the first time, some 3,5diglucosides. The investigated cultivars showed the same anthocyanin profile, but dramatic quantitative differences, i.e. the cultivar Sangiovese showed a lower amount of the acylated compounds. This analytical application was, therefore, useful as a supporting technique for the structural investigation Figure 8: Structure of the characteristic phenols of O. europea. of the polyphenolic compounds of different cultivars used in the production of red wines [40,41]. Numerous studies have been published on the analysis of olives, olive oil, waste waters (OMWW) and solid olive residue (SOR), experimental or commercial ones, from cultivars of different origins, stoned or whole fruits, and overall, there are more than 50 publications by the group up to now. However, in this case, I have selected some key studies carried out mainly by Prof. Nadia Mulinacci and Annalisa Romani, and more recently, by Prof. Patrizia Pinelli, as a graduate then PhD student, and as a postdoc. Olive oil is obtained from the olive (Olea europaea L., Oleaceae), a traditional tree crop of the Mediterranean Basin. On a European scale, 3 million tons of olives are processed for olive oil per year (with an oil yield of about 60,000 tons) [42]. After the epidemiological evidence of a lower incidence of CHD in the Mediterranean area [43] and certain types of cancers [44], there was an increasing popularity of the Mediterranean diet, in which olive oil is the major oil component, and its consumption is expanding to non producer countries such as the United States, Canada, and Japan. Olives and olive oil contain phenolic compounds, which not only influence the sensory properties, but are also important markers for type, biodiversity and quality determination of this product. These polyphenols have been shown to exert potent biological activities, including principally, but not limited to, antioxidant and free radical scavenging actions [42,45]. Some of the most representative phenolic compounds are hydroxytyrosol (3,4-dihydroxyphenylethanol), 1946 Natural Product Communications Vol. 3 (12) 2008 Bilia Antioxidant and anti-inflammatory properties were proven. A study on experimental and commercial OMWW from four Mediterranean countries (Italy, Spain, France, and Portugal) [52] was also carried out. The results demonstrated that Italian commercial OMWWs were the richest in total polyphenolic compounds with amounts between 150 and 400 mg/100 mL of waste waters. These raw, as yet unused, matrices were found to be an interesting and alternative source of biologically active polyphenols. Figure 9: Frantoio, a typical variety of Tuscan olive oil tyrosol, oleuropein, verbascoside and luteolin and its derivatives (Figure 8). Studies were first focused on the extraction of the minor polar compounds from olive fruit [46] and from extra virgin olive oils [47,48]. A solid-liquid extraction (LSE) procedure (Extrelut cartridge, diatomaceous earth), followed by HPLC-DAD-MS analysis led to the characterization of the polyphenolic content of different Tuscan olive cultivars (Frantoio, reported in Figure 9, Rossellino, Ciliegino, Cuoricino, and Grossolana), including phenolic acids, verbascoside, oleuropein derivatives, flavons and flavonol glycosides [46]. Verbascoside was proposed as a chemotaxonomic marker of different cultivars. Numerous comparative studies [47,48] were also carried out on extra-virgin oils from different parts of Italy and obtained from several harvest years (1999-2002) from both stoned and whole fruits. A higher antioxidant capacity of the oils from stoned olives was found. At the end of the ‘90s, a project supported by the European Community entitled "Natural antioxidants from olive oil processing waste waters" (FAIR PL 973039) was commenced with the aim of evaluating the polyphenolic contents in different samples of olive mill waste waters (OMWWs), and the possibility of recovering them. It is well-known that OMWWs contain powerful pollutants [49] which are acidic (pH 5-5.5) and malodorous, containing potassium and phosphorus salts and organic substances, such as fats, proteins, sugars, organic acids, but also polyphenols. For this purpose a preliminary qualitative screening of the polyphenols was performed working on waste waters obtained from an experimental mill in Tuscany [50], noting antioxidant and other biological activities of extracts obtained from this matrix [51]. Extracts presented both phenolic polymers and low and medium molecular weight phenols such as elenolic acid, hydroxytyrosol, and tyrosol. A further work was aimed at investigating the phenolic content of another by-product, the solid olive residue (SOR). The aim of this investigation was the selection of the best extraction procedure to increase the yields of phenylpropanoidic derivatives in the obtained extracts [53]. Soxhlet extraction with ethanol was identified as the first step of purification and was followed by either a liquid/liquid extraction with ethyl acetate or fractionation using an ionexchange resin. The total concentration of the phenolic compounds ranged between 1.1 and 6.23 mg/g of fresh SOR. Phenolic distribution among the different chemical classes was due to several factors: type of cultivar, degree of ripening, different milling processes, and pedoclimatic factors. The greatest differences among the samples were observed for verbascoside, which ranged between 0.15 and 4.15 mg g-1, and for its less abundant analogues [53]. When in 1997 I joined the group of Prof. Vincieri, my research studies were mostly directed toward pharmaceutical and technological aspects of herbal drugs, herbal drug preparations and herbal medicinal products. The first person I met in the Department was Dr. Sandra Gallori, an expert technician with a deep artistic sense. She was able to transform a simple poster into a work of art and the Vincieri group has been known for a long time in the scientific community also for these artistic posters, obtaining several awards. In many occasions she has prepared leaflets for congresses. An example of her work is a special picture representing the Vincieri group as an anthill (Figure 10). In 1998 Prof. Maria Camilla Bergonzi joined the group in order to start her PhD and during the following years, first as a post-doc and since 2005 as an aggregate professor, she has strongly supported my research. Authentication, quality control and stability testing have largely been performed using not only conventional [54-72] but also non conventional methods such as 1D- and 2D-NMR, NIR, and biosensors [73-77]. Franco F. Vincieri’s natural product research Natural Product Communications Vol. 3 (12) 2008 1947 S taff: Sandra Nadia Annalisa Anna Rita Patrizia Catia Vania Marzia Giovanni Camilla Carlotta Prof. Figure 10: A portrait of Vincieri research team. At the same time my scientific interest was driven by the improvement of bioavailability and technological features of extracts and HDPs, HMPs by formulation with liposomes, micelles, supramolecular complexes with cyclodextrins and their characterization by conventional methods (HPLC, DSC, UV, light scattering, dissolution tests) and innovative ones, such as NMR (ROESY and DOSY) [78-84]. Our publications represented the first studies on the stability and compatibility with excipients of important dried commercial extracts, such as St. John’s wort (SJW), and results were dramatic when the studies were carried out using ICH guidelines [56]. Other studies reported on the quality and/or stability of decoctions, teas and infusions, aromatic waters, tinctures and mother tinctures, commercial instant teas of common herbal drugs and obtained important information about their preparation, content and shelf-lives. Due to my long experience as a phytochemist and having at my disposal the excellent NMR facilities of Florence (CERM), one of the best equipped NMR groups in the world, I started a project using multidimensional NMR methods aimed at the analysis of complex spectra, such as those of plant extracts for their authentication, quality control and stability testing. Studies were performed by the direct NMR analysis of complex plant mixtures, without purification or fractionation steps. Many matrices were used, such as SJW, ginkgo and ginseng extracts, an innovative supercritical carbon dioxide (CO2) commercial extract of arnica, and samples of kava-kava herbal drugs and extracts [73-75]. Conventional methods used in herbal drug analysis (HPLC, HPTLC, GC, EC) can give a fingerprint of the markers or active constituents (and their percentage), but no information about the other metabolites of the extract, which can represent up to 95% of the total. This is true, especially in the unconventional extracts such as the innovative supercritical CO2 extracts. The extract or the finely powdered herbal drug were directly dissolved in hexadeuterated dimethylsulfoxide and analyzed after filtration by NMR spectroscopy. Spectral assignments of the constituents were carried out according to the data (chemical shifts and 1948 Natural Product Communications Vol. 3 (12) 2008 Bilia coupling constants) found in the literature and by means of 1D- and 2D-NMR spectra, which were found to be a valid alternative method to obtain a fingerprint for the assurance of content and stability and, as a consequence, safety and efficacy of extracts and herbal drugs. These studies represent a combination of fingerprint and semiquantitative analyses and although quantitation was only a minor aspect of these studies, the suitability of qHNMR to address questions of extract stability, as exemplified by the unstable Hypericum phloroglucinol derivatives, such as hyperforin, was clearly pointed out [56,73]. It was demonstrated that NMR experiments can provide a real and complete fingerprint of the extract, as required especially for innovative ones, to have a global vision and a “separation” of all constituents. All the molecules, as well as possible unknown or unexpected compounds can be detected. In addition, they can be used in the authentication of herbal drug material and to compare extracts manufactured with different processes and batch-to-batch analysis in the industry. NMR experiments can be considered a very simple, widely applicable and rapid analytical instrument, readily performed without pre-treatment, with tremendous versatility, not depending on the nature of the extract, inexpensive, perhaps with a lower precision than other methods but sufficient for pharmaceutical applications. Another important line of research was dedicated to the application of biosensors. This important application was possible thanks to collaboration with a colleague and dear friend, Prof. Maria Minunni, and her chief, Prof. Marco Mascini, of the Department of Chemistry of the University of Florence. Our studies proved the applicability of sensors and biosensors for analysis in the search for new active constituents from plants, for the quality-control of HDs, HDPs and HMPs related not only to active or marker constituents but also to other substances, such as heavy metals and pesticides. Different biosensors (based on electrochemical transduction or on optical detection) were employed to evaluate the content of alkaloids of different extracts of Chelidonium majus L. (Papavaraceae), fractions obtained during “biosensor assay-guided” fractionation, pure constituents and on extracts submitted for stability testing. In addition, disposable sensors were used to detect heavy metals in samples of St. John’s wort. A good correlation between the results obtained with the electrochemical devices and those from A.A.S. was observed [76,77]. Figure 11: Formation of liposome, a typical pharmaceutical carrier, in aqueous solutions. An important part of my research concerns the improvement of bioavailability and technological features of extracts, HDPs and HMPs by innovative methods (liposomes, ethosomes, supramolecular complexes, micelles) for which my thanks go also to Prof. Maria Camilla Bergonzi and many graduating and PhD students, among them Benedetta Isacchi, whom I consider to be my right arm. These preparations were analysed both by conventional methods (HPLC, DSC, UV, light scattering, dissolution tests) [78,80,81] and innovative ones, such as NMR (ROESY and DOSY) [79,82-84]. These studies were carried out on several supramolecular complexes between preparations of St. John’s wort, kava-kava and cyclodextrins, micelles and liposomes (Figure 11). I am really grateful to Prof. Gareth Morris of the University of Manchester (UK) for having applied and developed my initial idea of using diffusionordered spectroscopy (DOSY) methods for the analysis of micellar dispersions (octanoyl-6-Oascorbic acid, SDS) and included molecules, such as artemisinin, curcumin, phloroglucinols, and anthocyanins. The investigations showed that DOSY experiments can yield both qualitative and quantitative information on the solubilization of nonpolar species by surfactants and the supramolecular complexes. Finally, I should like to report some studies [85-89] with Artemisia annua L.. (Asteraceae, Figure 12) and artemisinin, a promising and potent antimalarial drug. These studies were carried out from the beginning in the framework of collaboration with a dear friend and colleague Prof. Luigi Messori of the Department of Analytical Chemistry of the University of Florence. Franco F. Vincieri’s natural product research Natural Product Communications Vol. 3 (12) 2008 1949 Artemisia annua L. Figure 12: Artemisia annua L. The interaction with hemin was first evaluated by UV/Vis spectrophotometry and HPLC/DAD/MS and the suitability of these simple methods for selection of new antimalarial compounds having similar properties was assessed. Furthermore the flavonoids isolated from A. annua were found to increase the rate of the reaction [85,86]. NMR studies of the supramolecular complex formed in the reaction were also carried out in collaboration with Prof. Paola Turano of the CERM [87]. Interesting results were also obtained using in the reaction hemoglobin instead of hemin [88]. Recent findings led to the discovery that green tea is also active against Plasmodium, and more importantly, its characteristic constituents, catechins, have a synergistic effect if administered with artemisinin [89]. Other studies on artemisinin and its extracts are also reported in this special issue. This project still represents one of the most visible for the group, involving many other scientists and dear friends including Dr Carlo Severini and Dr Anna Rosa Sannella of the Istituto Superiore di Sanità, Rome, Italy, and Prof. Deniz Tasdemir of the School of Pharmacy, University of London (UK), and also attracting funds from the Sigma Tau (Pomezia, Rome, Italy), Ente Cassa di Risparmio di Firenze (Florence, Italy) and Toscana Life Sciences Foundation (Siena, Italy). Recently, a collaborative study with Prof. Carla Ghelardini and Prof. Nicoletta Galeotti of the Department of Preclinical and Clinical Pharmacology of the University of Florence has begun, with the aim of investigating antineuropathic activity of extracts, fractions and pure compounds from herbal drugs (a paper concerning these studies is reported in this issue). Finally, last but not the least, I would like to remember Prof. Vincieri’s Figure 13: From the left side of the picture: Prof. K. Głowniak; Prof. T. Efferth, Prof. M. Hamburger, Prof. B.J. Baker, Prof. V. Butterweck, Prof. I. Merfort, Prof. L. Skaltsounis, Prof. R. Verpoorte, Prof. A. Nahrstedt, Prof. Prof. D. Guo, Prof. L. Pieters, Prof. A.R. Bilia and in the centre Prof. F.F. Vincieri on occasion of the celebration of his birthady during the 2008 Shangai International Conference on TCM and Natural Medicine. collaboration with Prof. Anacleto Minghetti and Dr. Nicoletta (Nicky) Crespi Perellino, wonderful people and outstanding scientists who started their sincere friendship with Franco from the first time they met. They are also very active people and their expertise lies in the field of cell cultures [90] in particular. The Vincieri research team is nowadays quite large, including three associate professors, three aggregate professors, four post-doctoral positions and about twenty students (including PhD, master’s and graduate students) having two principal lines of research. The first is related to the development of specific methods of extraction, fractionation, isolation and characterization of potentially interesting secondary metabolites for pharmaceutical, alimentary and cosmetic fields. Other studies are directed towards the optimization of stability and technological and biopharmaceutical characteristics of herbal products, their extracts and commercial preparations. I would like to conclude with a sentence to describe Prof Vincieri’s profile: “an inspration to scientists, young and old, in all fields of research”. Figure 13 is a photo of Prof. Vincieri in Shangai for the International Conference on Traditional Chinese Medicine and Natural Medicine taken on his birthday, 12th October 2008. At that time he had a nice surprise and I hope this issue will be another even a greater one! Happy birthday from all of us 1950 Natural Product Communications Vol. 3 (12) 2008 References [1] [2] [3] [4] [5] [6] [7] [8] [9] [10] [11] [12] [13] [14] [15] [16] [17] [18] [19] [20] [21] [22] [23] [24] [25] [26] [27] [28] Bilia ! Bambagiotti Alberti M, Vincieri FF. (1972) Monoterpene and sesquiterpene hydrocarbons of Pinus mugo. Phytochemistry, 11, 1455-1460. Bambagiotti Alberti M, Vincieri FF, Coran SA. (1972) Terpenoid hydrocarbons from Pinus pinea. Rivista Italiana Essenze, Profumi, Piante Officinali, Aromi, Saponi, Cosmetici, Aerosol, 54, 875-877. Bambagiotti Alberti M, Coran SA, Moneti G, Vincieri FF. (1978) Oxygenated compounds of Pinus montana Miller essential oil. Rivista Italiana Essenze, Profumi, Piante Officinali, Aromi, Saponi, Cosmetici, Aerosol, 60, 87-90. Bambagiotti Alberti M, Giannellini V, Coran SA, Vincieri FF. (1981) Infrared spectra for identification of naturally occurring bornyl, terpinenyl and alpha-terpinyl C3-C5 esters. Rivista Italiana Essenze, Profumi, Piante Officinali, Aromi, Saponi, Cosmetici, Aerosol, 63, 17-21. Bambagiotti Alberti M, Coran SA, Giannellini V, Vincieri FF, Moneti G. (1981) Inestigations of Pinus mugo essential oil oxygenated fraction by combined use of gas chromatography and dry column chromatography. Planta Medica, 43, 39-45. Bambagiotti Alberti M, Coran SA, Giannellini V, Moneti G, Vincieri FF, Selva A, Traldi P. (1981) Electron impact mass spectra of naturally occurring bornyl, 4-terpinenyl and alpha-terpinyl C3-C5 esters. Biomedical Mass Spectrometry, 8, 343-346. Bambagiotti Alberti M, Giannellini V, Coran SA, Vincieri FF, Moneti G, Selva A, Traldi P. (1982) Negative ion chemical ionization and collisional activation mass spectrometry of naturally occurring bornyl C4-C5 esters. Biomedical Mass Spectrometry, 9, 495-498. Bambagiotti Alberti M, Pinzauti S, Moneti G, Agati A, Giannellini V, Coran SA, Vincieri FF. (1984) Direct quantitation of antiseptic quaternary ammonium compounds by Fast Atom Bombardment Mass Spectrometry. Journal of Pharmaceutical and Biomedical Analysis, 2, 409-415. Bambagiotti Alberti M, Pinzauti S, Vincieri FF. (1987) Simultaneous capillary gas chromatographic determination of guaifenesin, dextromethorphan and diphenhydramine in cough-cold syrup. Pharmaceutica Acta Helvetiae, 62, 5-6. Vincenzini MT, Vincieri FF, Vanni P. (1973) The effect of octanoate and oleate on isocitrate lyase activity during the germination of Pinus pinea seeds. Plant Physiology, 52, 549-553. Vincenzini MT, Vincieri FF, Vanni P. (1975) In vivo effect of octanoate and oleate on glucose-6-phosphate dehydrogenase during germination of Pinus pinea seeds. The Italian Journal of Biochemistry, 24, 317-324. Vanni P, Vincenzini MT, Vincieri FF. (1975) The composition and utilization of lipids and the operation of the glyoxylate cycle during the germination of Lupinus seeds. Experientia, 31, 1392-1394. Vanni P, Vincieri FF, Vincenzini MT. (1976) Sensitivity of glucose-6-phosphate dehydrogenase activity to saturated and unsaturated fatty acids. Canadian Journal of Biochemistry, 54, 760-764. Vanni P, Vincenzini MT, Vincieri FF, Baccari V. (1977) Stimulation of isocitrate lyase biosynthesis by hydroxylamine and hydrazine. Molecular and Cellular Biochemistry, 15, 125-131. Vincenzini MT, Nerozzi F, Vincieri FF, Vanni P. (1980) Isolation and properties of isocitrate lyase from Lupinus seeds. Phytochemistry, 19, 769-774. Vincieri FF, Vincenzini MT, Vanni P. (1981) Extraction of active compounds from sarcotesta of Ginkgo biloba seeds: inhibition of some dehydrogenases activity. Rivista Italiana Essenze, Profumi, Piante Officinali, Aromi, Saponi, Cosmetici, Aerosol, 63, 79-82. Rzadkowska-Bodalska H. (1994) Sedum telephium. In "Hagers Handbuch der Pharmazeutischen Praxis", 5th edn., Vol. 5, Hänsel R, Keller K, Rimpler H, Schneider G. (Eds). Springer-Verlag, Berlin, Heidelberg, New York., 650-655. Balatri S. (1981) L'uso del Sedum telephium L. nella pratica chirurgica minore. Bollettino della Società Tosco-Umbra di Chirurgia XLII, 3-8. Vincieri FF, Mulinacci N, Coran SA, Bambagiotti Alberti M. (1985) Approccio allo studio dei principi attivi del Sedum telephium. Studi Sassaresi, 45, 21-25. Sendl A, Mulinacci N, Vincieri FF, Wagner H. (1993) Anti-inflammatory and immunologically active polysaccharides of Sedum telephium. Phytochemistry, 34, 1357-1362. Bambagiotti Alberti M, Sendl A, Wagner H, Mulinacci N, Vincieri FF, Baldi A. (1995) Flavonol glycosides from Sedum telephium subspecies maximum leaves. Phytochemistry, 38, 531-533. Mulinacci N, Vincieri FF, Baldi A, Romani A, Faretto D, Traldi P. (1995) Mass spectrometric methodologies in plant analysis: The case of flavonols in Sedum telephium L. juice Rapid Communications in Mass Spectrometry, 9, 963-967. Sturm S, Mulinacci N, Vincieri FF, Stuppner H. (1999) Analysis of flavonols of Sedum telephium L. leaves by capillary electrophoresis and HPLC-mass spectrometry. Chromatographia, 50, 433-438. Bonina F, Puglia C, Tomaino A, Saija A, Mulinacci N, Romani A, Vincieri FF. (2000) In vitro antioxidant and in vivo photoprotective effect of three lyophilized extracts of Sedum telephium L. leaves. Journal of Pharmacy and Pharmacology, 52, 1271-1277. Raimondi L, Banchelli G, Dalmazzi D, Mulinacci N, Romani A, Vincieri FF, Pirisino R. (2000) Sedum telephium L. polysaccharide content affects MRC5 cell adhesion to laminin and fibronectin. Journal of Pharmacy and Pharmacology, 52, 585-591. Grieve M. (1967) Dropwort, Hemlock Water. In A Modern Herbal The Medicinal, Culinary, Cosmetic and Economic Properties, Cultivation and Folk-Lore of Herbs, Grasses, Fungi, Shrubs & Trees with their Modern Scientific Uses . Vol. 1, Leyel CF (Ed), Hafner Publishing Co., New York. 264-272. Watson W. (1757/1758) A further account of the poisonous effects of the Oenanthe aquatica succo viroso crocante of Lobel, or hemlock dropwort. Philosophical Transactions (1683-1775), 50, 856-859. Vincieri FF, Coran SA, Bambagiotti Alberti M. (1976) Composition of the Oenanthe aquatica essential oil. Planta Medica, 29, 101-112. Franco F. Vincieri’s natural product research [29] [30] [31] [32] [33] [34] [35] [36] [37] [38] [39] [40] [41] [42] [43] [44] [45] [46] [47] [48] [49] [50] [51] [52] [53] [54] [55] [56] [57] [58] [59] Natural Product Communications Vol. 3 (12) 2008 1951 Vincieri FF, Coran SA, Giannellini V, Bambagiotti Alberti M. (1981) Isolation and structural elucidation of Oenanthe aquatica L fruit C15 polyacetylene hydrocarbons. Chemische Berichte, 114, 468-476. Vincieri FF, Coran SA, Giannellini V, Bambagiotti Alberti M. (1985) Oxygenated C15polyacetylenes from Oenanthe aquatica L fruit. Planta Medica, 107-110. Vincieri FF, Coran SA, Bambagiotti Alberti M, Smulevich G, Marzocchi MP. (1986) Second-derivative UV spectra of polyacetylene chromophores: fingerprints of their geometrical isomers. Chemische Berichte, 119, 2843-2847. Smulevich G, Marzocchi MP, Vincieri FF, Coran SA, Bambagiotti Alberti M. (1987). Vibronic absorption spectra of naturally occurring conjugated polyenynes. Evidence for localized excitations. Journal of Chemical Society, Perkin Transactions, 1431-1437. Coran SA, Bambagiotti alberti M, Vincieri FF, Moneti G. (1987) Rapid monitoring of biologically-active substances in medicinal plants by tandem mass spectrometry. The identification of lignans in Oenanthe aquatica Lam. Journal of Pharmaceutical and Biomedical Analysis, 5, 509-514. Vincieri FF, Coran SA, Mulinacci N, Bambagiotti Alberti M. (1989) An insight into Oenanthe aquatica L fruit tincture through the MS identification of its gas chromatographable compounds. Pharmaceutica Acta Helvetiae, 64, 30-34. Vincieri FF, Mulinacci N, Mazzi G, Coran SA, Bambagiotti Alberti M. (1988) Stabilization of tincture with cyclodextrins: a tincture from the fruits of Oenanthe aquatica L. International Journal of Pharmaceutics, 48, 119-123. Vincieri FF, Mazzi G, Mulinacci N. (1994) Permeation of Oenanthe aquatica fruit tincture-β-cyclodextrin complex through artificial membranes. Il Farmaco, 49, 63-67. Romani A, Pinelli P, Mulinacci N, Vincieri FF, Gravano E, Tattini M. (2000) Identification and quantitation of flavonoids and secoiridoids in leaves of Ligustrum vulgare L. (Oleaceae). Journal of Agricultural and Food Chemistry, 48, 4091-4096. Pinelli P, Romani A, Mulinacci N, Galardi C, Vincieri FF, Tattini M. (2000) HPLC analysis of polyphenols in Pistacia lentiscus L. leaves. Polyphenols 2000, INRA editions, Paris, France, 78-83. Romani A, Coinu R, Carta S, Pinelli P, Galardi C, Vincieri FF, Franconi F. (2004) Evaluation of antioxidant effect of different extracts of Myrtus communis L. Free Radical Research, 38, 97-103. Baldi A, Romani A, Mulinacci N, Vincieri FF, Casetta B. (1995) HPLC/MS application to anthocyanins of Vitis vinifera L. Journal of Agricultural and Food Chemistry, 43, 2104-2109. Baldi A, Romani A, Mulinacci N, Vincieri FF. (1993) Polyphenols in Tuscan varieties of Vitis vinifera L. Journal International des Sciences de la Vigne et du Vin, 27, 201-215. Visioli E, Galli C (1998) Olive oil phenols and their potential effects on human health. Journal of Agricultural and Food Chemistry, 46, 4292-4296. Hertog MGL, Kromhout D, Aravanis C, Blackburn H, Buzina R, Fidanza F, Giampaoli S, Jansen A, Menotti A, Nedeljkovic S, Pekkarinen M, Simic BS, Toshima H, Feskens EJM, Hollman PCH, Katan MB. (1995) Flavonoid intake and long-term risk of coronary heart disease and cancer in the Seven Countries Study. Archives of Internal Medicine, 155, 381-386. Lipworth L, Martinez ME, Angell J, Hsien CC, Trichopoulos D. (1997) Olive oil and human cancer an assessment of the evidence. Preventive Medicine, 26, 181-190. Visioli F, Galli C. (1998) The effect of minor constituents of olive oil on cardiovascular disease: new findings. Nutrition Reviews, 56, 142-147. Romani A, Mulinacci N, Pinelli P, Vincieri FF, Cimato, A. (1999) Polyphenolic content in five tuscany cultivars of Olea europaea L. Journal of Agricultural and Food Chemistry, 47, 964-967. Romani A, Pinelli P, Mulinacci N, Galardi C, Vincieri FF, Liberatore L, Cichelli A. (2001) HPLC and HRGC analysis of polyphenols and secoiridoids in olive oil. Chromatographia, 53, 279-284. Mulinacci N, Giaccherini C, Innocenti M, Romani A, Vincieri FF, Marotta F, Mattei A. (2005) Analysis of extra virgin olive oils from stoned olives. Journal of the Science of Food and Agriculture, 85, 662-670. Saviozzi A, Riffaldi R, Levi-Minzi R, Scagnozzi A, Venni G. (1993). Decomposition of vegetation-water sludge in soil. Bioresource Technology, 44, 223-228. Romani A, Baldi A, Mulinacci N, Vincieri FF, Mancini P. (1996). Acque reflue di frantoio: studio dei composti polifenolici per un eventuale uso alternativo. Problemi e prospettive. Atti del XVII Congresso Nazionale di Merceologia. Merci e cicli produttivi nel settore agroindustriale alle soglie del 21secolo, 1, 834-841. Visioli F, Romani A, Mulinacci N, Zarini S, Conte D, Vincieri FF, Galli C. (1999) Antioxidant and other biological activities of olive mill waste waters. Journal of Agricultural and Food Chemistry, 47, 3397-3401. Mulinacci N, Romani A, Galardi C, Pinelli P, Giaccherini C, Vincieri FF. (2001) Polyphenolic content in olive oil waste waters and related olive samples. Journal of Agricultural and Food Chemistry, 49, 3509-3514. Mulinacci N, Innocenti M, la Marca G, Mercalli E, Giaccherini C, Romani A, Erica S, Vincieri FF. (2005) Solid olive residue: an insight on their polyphenolic composition. Journal of Agricultural and Food Chemistry, 53, 8963-8969. Bilia AR, Fumarola M, Gallori S, Mazzi G, Vincieri FF. (2000) Identification by HPLC-DAD and HPLC-MS analyses and quantification of constituents of fennel teas and decoctions. Journal of Agricultural and Food Chemistry, 48, 4734-4738. Bilia AR, Salvini D, Mazzi G, Vincieri FF. (2001) Characterization of calendula, milk-thistle and passionflower tinctures by HPLC-DAD and HPLC-MS. Chromatographia, 53, 210-215. Bilia AR, Bergonzi MC, Morgenni F, Mazzi G, Vincieri FF. (2001) Evaluation of stability of St. John’s wort commercial extract and some preparations. International Journal of Pharmaceutics, 213, 199-208. Bergonzi MC, Bilia AR, Gallori S, Guerrini D, Vincieri FF. (2001) Variability in the content of the constituents of Hypericum perforatum and some commercial extracts. Drug Development and Industrial Pharmacy, 27, 491-97. Gallori S, Flamini G, Bilia AR, Morelli I, Landini A, Vincieri FF. (2001) Chemical composition of some traditional herbal drug preparations: essential oil and aromatic water of Costmary (Balsamita suaveolens Pers.). Journal of Agricultural and Food Chemistry, 49, 5907-5910. Bilia AR, Flamini G, Taglioli V, Morelli I, Vincieri FF. (2002) GC-MS analysis of essential oil of some commercial Fennel teas. Food Chemistry, 76, 307-310. 1952 Natural Product Communications Vol. 3 (12) 2008 [60] [61] [62] [63] [64] [65] [66] [67] [68] [69] [70] [71] [72] [73] [74] [75] [76] [77] [78] [79] [80] [81] [82] [83] [84] [85] [86] [87] [88] [89] [90] Bilia Bilia AR, Bergonzi MC, Mazzi G, Vincieri FF. (2002) HPLC-DAD and HPLC-MS analysis and stability of the constituents of Artichoke and St. John’s wort tinctures. Drug Development and Industrial Pharmacy, 28, 609-619 Bilia AR, Bergonzi MC, Mazzi G, Vincieri FF. (2002) Stability of the constituents of Calendula, Milk-thistle and Passionflower tinctures by HPLC-DAD and HPLC-MS analyses. Journal of Pharmaceutical and Biomedical Analysis, 30, 613-617. Sturm S, Taglioli V, Bilia AR, Vincieri FF, Stuppner H. (2003) Analysis of alkaloids in Ipecacuanhae radix and preparations by capillary zone electrophoresis. Journal of Separation Science, 26, 1175-1179. Gallori S, Bilia AR, Bergonzi MC, Barbosa WLR, Vincieri.FF. (2004) Polyphenolic constituents of fruit juice of Euterpe oleracea Mart. (Açai palm). Chromatographia, 59, 739-743. Gallori G, Giaccherini C, Bilia AR, Mulinacci N, Vincieri FF. (2004) Identification of anthocyanins in Amorphophallus titanum (Becc.) Becc.. Chromatographia, 59, 647-651. Bilia AR, Melillo de Malgalhaes P, Bergonzi MC, Vincieri FF. (2006) Simultaneous analysis of artemisinin and flavonoids of several extracts of Artemisia annua L. obtained from a commercial sample and a selected cultivar. Phytomedicine, 13, 487-493. Karioti, A, Chatzopoulou A, Bilia AR, Liakopoulos G, Stavrianakou S, Skaltsa S. (2006) Novel secoiridoid glucosides in Olea europaea leaves suffering from boron deficiency. Bioscience, Biotechnology and Biochemistry, 70, 1898-1903. Bilia AR, Gabriele C, Bergonzi MC, Melillo de Malgalhaes P, Vincieri FF. (2006) Variation in artemisinin and flavonoids content in different extracts of Artemisia annua L. Natural Product Communications, 1, 1111-1115. Bilia AR, Eterno F, Bergonzi MC, Mazzi G, Vincieri FF. (2007) Evaluation of the content and stability of the constituents of mother tinctures and tinctures: the case of Crataegus oxyacantha L. and Hieracium pilosella L.. Journal of Pharmaceutical and Biomedical Analysis, 44, 70-78. Isacchi B, Bergonzi MC, Carnevali F, van der Esch SA, Vincieri FF, Bilia AR. (2007) Analysis and stability of the constituents of St. John's wort oils prepared with different methods. Journal of Pharmaceutical and Biomedical Analysis, 45, 756-761. Baraldi R, Isacchi B, Predieri S, Marconi G, Vincieri FF, Bilia AR. (2008) Distribution of artemisinin and bioactive flavonoids from Artemisia annua L. during plant growth. Biochemical Systematics and Ecology, 36, 340-348. Bilia AR, Giomi M, Innocenti M, Gallori S, Vincieri FF. (2008) HPLC-DAD-ESI-MS analysis of the constituents of aqueous preparations of verbena and lemon verbena and evaluation of the antioxidant activity. Journal of Pharmaceutical and Biomedical Analysis, 46, 463-470. Birilli R, Ferretti R, Gallinella B, Bilia AR, Vincieri FF, La Torre F. (2008) Enantioseparation of kavain on Chiralpak IA under normal-phase, polar organic and reversed-phase conditions. Journal of Separation Science, 31, 2206-2210. Bilia AR, Bergonzi MC, Mazzi G, Vincieri FF. (2001) Analysis of plant complex matrices by use of nuclear magnetic resonance spectroscopy: St. John’s wort extract. Journal of Agricultural and Food Chemistry, 49, 2115-2124. Bilia AR, Bergonzi MC, Lazari D, Vincieri FF. (2002) Characterisation of commercial kava-kava herbal drug and herbal drug preparations by means of nuclear magnetic resonance spectroscopy. Journal of Agricultural and Food Chemistry, 50, 5016-5025. Bilia AR, Bergonzi MC, Mazzi G, Vincieri FF. (2002) Analysis of plant complex matrices by use of nuclear magnetic resonance spectroscopy: supercritical carbon dioxide arnica extract. Journal of Pharmaceutical and Biomedical Analysis, 30, 321-330. Palchetti I, Mascini M, Minunni M, Bilia AR, Vincieri FF. (2003) Disposable electrochemical sensor for rapid determination of heavy metals in herbal drugs. Journal of Pharmaceutical and Biomedical Analysis, 32, 251-256. Minunni M, Tombelli S, Marrazza G, Palchetti I, Mascini M, Bilia AR, Bergonzi MC, Vincieri FF. (2005) An optical DNA-based biosensor for the analysis of bioactive constituents with application in drug and herbal drug screening. Talanta, 65, 578-585. Taglioli V, Bilia AR, Ghiara C, Mazzi G, Mercati V, Vincieri FF. (2001) Evaluation of the dissolution behaviours of some commercial herbal drugs and their preparations. Pharmazie, 56, 868-870. Bilia AR, Bergonzi MC, Morris GA, Lo Nostro P, Vincieri FF. (2002) A diffusion-ordered NMR spectroscopy study of the solubilization of artemisinin by octanoyl-6-O-ascorbic acid. Journal of Pharmaceutical Sciences, 91, 2265-2270. Bergonzi MC, Bilia AR, Casiraghi A, Cilurzo F, Minghetti P, Montanari L, Vincieri FF. (2005) Evaluation of skin permeability of sesquiterpenes of an innovative supercritical carbon dioxide arnica extract by HPLC/DAD/MS. Die Pharmazie, 60, 36-38. Bilia AR, Bergonzi MC, Mazzi G, Vincieri FF. (2006) Development and stability of semisolid preparations based on a supercritical CO2 arnica extract. Journal of Pharmaceutical and Biomedical Analysis, 41, 449-54. Bergonzi MC, Bilia AR, Di Bari L, Mazzi G, Vincieri FF. (2007) Studies on the interactions between some flavonols and cyclodextrins. Bioorganic & Medicinal Chemistry Letters, 17, 5744-5748. Sinico C, Caddeo C, Valenti D, Fadda AM, Bilia AR, Vincieri FF. (2008) Liposomes as carriers for verbascoside: stability and skin permeation studies. Journal of Liposome Research, 18, 83-90 Bilia AR, Scalise L, Bergonzi MC, Vincieri FF. (2008) Effect of surfactants and solutes (glucose and NaCl) on solubility of kavain. American Association of Pharmaceutical Scientists-(AAPS Pharmscitech), 9, 444-448. Bilia AR, Lazari D, Messori L, Taglioli V, Temperini C, Vincieri FF. (2002) Simple and rapid physico-chemical methods to examine action of antiimalarial drugs: its application to Artemisia annua L. constituents. Life Sciences, 70, 769-778. Messori L, Piccioli F, Eitler B, Bergonzi MC, Bilia AR, Vincieri FF. (2003) Spectrophotometric and ESI-MS/HPLC studies reveal a common mechanism for the reaction of various artemisinin analogues with hemin. Bioorganic and Medicinal Chemistry letters, 13, 4055-4057. Messori L, Piccioli F, Temperini C, Bilia AR, Vincieri FF, Allegrozzi M, Turano P. (2004) The reaction of artemisinin with hemin: a further insight into the mechanism. Inorganica Chimica Acta, 357, 4602-4606. Messori L, Gabbiani C, Casini A, Siragusa M, Vincieri FF, Bilia AR. (2006) The reaction of artemisinins with hemoglobin: A unified picture. Bioorganic & Medicinal Chemistry, 14, 2972-2977. Sannella AR, Messori L., Casini A, Vincieri FF, Bilia AR, Majori G, Severini C. (2007) Antimalarial properties of green tea. Biochemical and Biophysical Research Communications, 353, 177-181. Mulinacci N, la Marca G, Innocenti M, Vincieri FF, Crespi Perellino N, Minghetti A. (2005) Cell cultures of Ajuga reptans L. to bioconvert emodin and aloe-emodin: an HPLC/ESI/MS investigation. Enzyme and Microbial Technology, 36, 399-408. NPC Natural Product Communications Effects of Terpenoids from Salvia willeana in Delayed-type Hypersensitivity, Human Lymphocyte Proliferation and Cytokine Production 2008 Vol. 3 No. 12 1953 - 1958 Anna Vonapartib, Anastasia Kariotib, María C. Recioa, Salvador Máñeza, José L. Ríosa, Eleani Skaltsab and Rosa M. Ginera,* a Departament de Farmacologia, Facultat de Farmàcia, Universitat de València, Av. Vicent Andrés Estellés s/n, 46100 Burjassot, Spain b Department of Pharmacognosy and Chemistry of Natural Products, School of Pharmacy, University of Athens, Panepistimiopolis, Zografou 157 71, Athens, Greece Rosa.M.Giner@uv.es Received: June 24th, 2008; Accepted: October 16th, 2008 The effect of the lipophilic extract of S. willeana and three terpenoids isolated therefrom, camphor, lupeol and oleanolic acid, on oxazolone-induced hypersensitivity was evaluated. The extract reduced the ear edema by 46% at 24 h after challenge. All three terpenoids inhibited the edema and suppressed cytokines release at different rates. Lupeol inhibited the swelling by over 50% and reduced the production of IL-1β by 62%. Camphor caused inhibition of the efferent phase (45% inhibition at 72 h) and the levels of IL-1β, IL-4 and TNF-α (around 80% inhibition). Oleanolic acid diminished moderately the reaction and the levels of IL-4 and TNF-α. We also demonstrated that the three terpenoids inhibited human T-lymphocytes proliferation in a concentration-dependent manner and induced their apoptosis. Thus, these terpenoids could be considered anti-inflammatory constituents of S. willeana. Keywords: Salvia willeana, oleanolic acid, lupeol, camphor, oxazolone, lymphocytes, cytokines. The genus Salvia comprises about 900 species, many of which are used in traditional medicine to treat various disorders [1,2]. S. willeana (Holmboe) Hedge (Lamiaceae), an aromatic herb endemic to Cyprus, and locally common in the Troodos range [3], possesses pharmacological properties. Its extracts or infusions are used as tonics and anti-diarrhoeal agents. The plant can also be used to halt milk production in nursing mothers and has a strong antiseptic action [4,5]. This species has been characterized by the presence of the triterpenoids, ursolic and oleanolic acids and urs-12-ene-3α,11αdiol, the diterpenoids, carnosic acid and isorosmanol and the flavonoid salvigenin [6]. These classes of compounds have demonstrated anti-inflammatory activity in different experimental models of inflammation. In the present study we report the activity of the lipophilic extract of the aerial parts of S. willeana and the three isolated compounds, camphor, lupeol and oleanolic acid, on a delayed-type hypersensitivity reaction induced by oxazolone, specifically on edema formation and their effect on different proinflammatory mediators involved in this reaction, as well as their influence on both T lymphocyte proliferation and the cell cycle. The lipophilic extract, which was topically applied at a dose of 1 mg/ear after oxazolone challenge, reduced the ear edema by 46% at 24 h, but exhibited only a weak effect during the later stages of the process (22% and 20% at 48 h and 72 h, respectively). When topically applied at a dose of 0.5 mg/ear, the isolated compounds clearly and significantly reduced the hypersensitivity reaction in mouse ears (Figure 1). Lupeol was the most active compound, inhibiting the swelling by 58% 24 h after challenge and maintaining the effect after 72 h (50% and 52% at 48 and 72 h, respectively). Camphor caused a slight 1954 Natural Product Communications Vol. 3 (12) 2008 Vonaparti et al. Control 1000 175 Lupeol 250 800 ** Oleanolic acid 150 ** 125 ** ** 100 Dexamethasone ** 75 50 ** 25 ** ** TNF-α and IL-4 (pg/mL) 200 300 200 600 150 100 ** 50 ** 400 ** ** ** 10 20 30 40 50 60 70 Time (h) Figure 1: Effect of the extract, the isolated compounds and dexamethasone on the delayed-type hypersensitivity ear swelling induced by oxazolone, measured 24, 48 and 72 h after challenge. Increase in ear thickness is expressed as mean ± SEM. ** P < 0.01 after Dunnett’s test as compared with control group. inhibition of the elicitation phase (28% at 24 h), but its effect intensified at 72 h (45% inhibition), whereas oleanolic acid produced a moderate reduction of the reaction (37%, 33%, and 30% at 24 h, 48 h, and 72 h, respectively) (Figure 1). A critical event during the development of cutaneous immune responses, including those provoked by exposure to a contact allergen such as oxazolone, is the mobilization of epidermal Langerhans cells (LC). These cells act as sentinels of the immune system in the skin, responding to a variety of local injuries with migration and the delivery of potentially foreign signals leading to the draining of the lymph nodes. IL-1β and TNF-α are known to play pivotal roles in the stimulation of LC migration. Thus, with regard to the effect of the compounds on the protein level of such cytokines, IL-1β, TNF-α, and IL-4 production were measured in ear homogenates 72 h after challenge with oxazolone with the aid of an ELISA analysis. The effectiveness of the compounds varied depending on the cytokine, with the inhibitory activity on the liberation of all three cytokines being especially marked for camphor (around 80% inhibition with respect to those in the acetone-treated group). In contrast, the other compounds exhibited different behaviors. Oleanolic acid, for example reduced the levels of IL-4 and TNF-α by 42% and 46%, respectively, but did not modify the concentration of IL-1β. Lupeol notably reduced the production of IL-1β by 62%, moderately reduced that of IL-4 by 36%, but had no significant effect on TNF-α levels. In the dexamethasone-treated group, a decrease in the production of cytokines to basal levels was observed (Figure 2). 0 Bl an 0 200 IL-1β k Co nt ro l Ca m ph or Lu pe O le ol an ol i c D ac ex id am et ha so ne 0 0 ** IL-4 IL-1β (pg/mL) ΔEar swelling (μm ± SEM) TNF-α Extract Camphor 225 Figure 2: Effect of isolated compounds and dexamethasone on IL-1β, TNF-α and IL-4 production in ear homogenates. Data represent mean ± SEM of at least three independent experiments. ** P < 0.01 after Dunnett’s test, as compared with control group. The study of cell viability and toxicity, assessed by examining the mitochondrial reduction of MTT after 24 h, showed that at 100 μM, none of the tested compounds was toxic to either human lymphocytes or murine RAW 264.7 macrophages (data not shown). In LPS-stimulated macrophages, lupeol at a final concentration of 100 μM reduced the nitrite production in the culture medium by 55% while camphor only exhibited a slight inhibition (25%). These results indicate an effect on the production of nitric oxide, which plays a relevant role in contact dermatitis and contributes to the swelling and infiltration of effector cells. All three tested compounds, camphor, lupeol, and oleanolic acid, inhibited T-cell proliferation in a concentration dependent manner 72 h after PHA stimulation, with IC50 values of 3.7, 1.6, and 3.3 μM, respectively. We then set out to determine the phases in which the cell cycle was modified. After incubation, either with or without the compounds, the cell cycle was analyzed with propidium iodide reagent and subsequent flow cytometry analysis. The analysis of the cell cycle at different times indicated that resting T-cells stayed mainly in the G0/G1 phase, whereas PHA-stimulated cells went from the G0/G1 phase to the S phase and then on to the G2/M phase. When PHA-stimulated cells were treated with the isolated compounds, the passage to the S and M phases was reduced, with the maximum effect appearing 72 h after stimulation. These compounds actually induced the apoptosis of human lymphocytes, with the percentage of cells in the subG0 phase higher at 24 and 72 h with respect to the control group (Figure 3). Effects of terpenoids of Salvia willeana on hypersensitivity Natural Product Communications Vol. 3 (12) 2008 1955 A) edema, and lupeol was active against 12deoxyphorbol-13-phenylacetate- and bryostatin-1induced edemas. These findings indicate that the inhibition of protein kinase C plays a role in their anti-inflammatory mechanism [7]. Oral administration of lupeol inhibited CD4+ and CD8+ T cells as well as cytokines IL-2, IL-4, and IFN-γ in a DTH reaction induced by ovalbumin in mice [8]. Oleanolic acid exhibited moderate activity on the DTH reaction induced by dinitrofluorobenzene, suppressing the edema by 32% 96 h after challenge [9]. This compound also enhanced the total white blood cell count [10] and increased the total antibody production in the same way as the monoterpenes carvone, limonene, and perillic acid, indicating its potentiating effect on the immune system [11]. However, due to their irritant properties, camphor and limonene were unable to induce an immunostimulatory response in the popliteal lymph node assay in rats [12]. A case report of an allergic contact dermatitis from rectified camphor oil as a component of a topical medicine has been previously published [13]. Still, it has been reported that T cell activation, proliferation, and cytokine gene transcription are all regulated by several transcription factors in which NF-κB shows a significant participation. In this context, in a search for inhibitory natural products from medicinal plants against NF-AT transcription factor, it was found that oleanolic acid showed an IC50 > 50 μM [14]. 100 Percentage ± SEM Sub G0 Go 75 S M 50 25 c A PH A + O PH A + le an ol ic Lu pe ol ph or C am PH A PH A + B la nk 0 B) Percentage ± SEM 90 80 Sub G0 70 Go 60 S 50 M 40 30 20 10 A c O le an ol ic Lu pe ol + PH A PH A PH + A C + am ph or A PH B la nk 0 C) 100 Percentage ± SEM Sub G0 Go 75 S M 50 25 A c O le an ol ic Lu pe ol + PH A PH A C + A PH + am ph or A PH B la nk 0 Figure 3: Effect of isolated compounds on the cell cycle phases at 12 h (A), 24 h (B) and 72 h (C) after PHA-stimulation. Time is expressed as hours after addition of 8 μM isolated compounds subsequent to PHA stimulation. Values represent the percentage of cells in each phase of the cell cycle ± SEM. A number of triterpenoids, including oleanolic acid and lupeol, exhibit marked anti-inflammatory activity and have been found to modulate the immune system. We found both triterpenoids to be effective when applied topically against various protein kinsase C activators. For example, both inhibited 12deoxyphorbol-13-tetradecanoate-induced edema, while oleanolic acid reduced merezein-induced Considering the variety of bioactive triterpenoids from nearly 100 Salvia species reported in the literature [15], it seems clear that this kind of compound is fairly representative within the genus. So, taking into account the results obtained for the terpenoids assayed in this study, we must assume that the pharmacological properties attributed to Salvia willeana correlate with the occurrence of these constituents, which support its use in traditional medicine and which could be extended to other species of Salvia with the same constituents. In conclusion, this study demonstrates that S. willeana possesses anti-inflammatory activity and that lupeol, oleanolic acid, and camphor could be considered as a part of its active constituents. They are most likely responsible for the plant’s potential therapeutic benefits since they attenuate the inflammatory reaction induced by oxazolone and exert a proliferation-suppressive action, in part through a reduction in the release of inflammatory cytokines. 1956 Natural Product Communications Vol. 3 (12) 2008 Experimental General experimental procedures: 1H, 13C and 2D NMR spectra were recorded in CDCl3 on Bruker DRX 400 and Bruker AC 200 (50.3 MHz for 13C NMR) instruments at 295° K. Chemical shifts are given in ppm (δ) and were referenced to the solvent signals at δ 7.24 and 77.0 for 1H and 13C NMR, respectively. IR spectra were obtained on a PerkinElmer PARAGON 500 FT-IR spectrophotometer. The [α]D values were obtained in CDCl3 at 20ºC on a Perkin - Elmer 341 polarimeter. Vacuum liquid chromatography: silica gel 60H (Merck); Column chromatography (CC): silica gel 60 (SDS, 40-63 μm), gradient elution with the solvent mixtures indicated in each case; Sephadex LH-20 (Pharmacia) with MeOH; cyclohexane-CH2Cl2-MeOH. TLC: Merck silica gel 60 F254. Detection: UV-light, spray reagent (vanillin-H2SO4; anisaldehyde-H2SO4). Plant material: Aerial parts of Salvia willeana (Holmboe) Hedge were collected on Troodos Mountain (Cyprus), in April 2004. A voucher specimen has been deposited in the Agricultural Research Institute Herbarium of Nicosia [nº ARI 3213]. Extraction and isolation: The air-dried powdered aerial parts of S. willeana (0.43 kg) were successively extracted at room temperature with cyclohexane, dichloromethane, MeOH and MeOH/H2O (5/1) (2 L of each solvent, twice for 48 h). The combined dried cyclohexane and dichloromethane extracts (45.0 g) were subjected to vacuum liquid chromatography (VLC) over silica gel (8 x 6.0 cm) with cyclohexane/CH2Cl2 (90:10-10:90); CH2Cl2-MeOH mixtures (99:1-80:20) of increasing polarity to yield eighteen fractions (A-R) of 500 mL. Fraction F (0.36 g, eluted with cyclohexane/CH2Cl2 50:50), was identified as camphor (1). Fraction H (0.80 g, eluted with cyclohexane/CH2Cl2 30:70) was further subjected to repeated CC over silica gel using cyclohexane-EtOAc mixtures of increasing polarity (99:1-96:4) and Sephadex LH-20 (cyclohexane/CH2Cl2/MeOH 7:4:0.2) and afforded lupeol (2; 30.5 mg). Fraction N (3.0 g, eluted with CH2Cl2-MeOH 97:3) was further purified on Sephadex LH 20 (CH2Cl2-MeOH mixtures of increasing polarity) yielding oleanolic acid (3; 31.0 mg). These compounds were identified by comparing their chromatographic and spectroscopic data with those of pure standards. Vonaparti et al. Oxazolone-induced delayed-type hypersensitivity [16]: Female CD-1 mice (Harlan Interfauna Iberica, Barcelona, Spain), weighing 25–30 g, were randomly distributed into groups of 6 animals and fed with a standard diet and water ad libitum. Housing conditions and the protocol were approved by the Ethical Committee of the Faculty of Pharmacy in accordance with the guidelines established by the European Union on Animal Care (CEE Council 86/609). At the beginning of the experiment, on day 1, mice were sensitized by means of topical application to the shaved abdomen of 150 μL of a 3% (w/v) solution of oxazolone in acetone. On day 2, ear thicknesses were measured to obtain data for ears with no inflammation. On day 6, challenge was performed by applying 20 μL of 1% (w/v) oxazolone in acetone to both the inner and outer surfaces of both ears, after which the extract (1 mg/ear) and test compounds (compounds 1, 2 and 3 at 0.5 mg/ear and dexamethasone 0.025 mg/ear), dissolved in either acetone or EtOH/H2O (7:3) were applied topically (20 μL) to the ears either 1, 24 or 48 h after challenge. Ear thickness of both the treated and control groups was measured with a micrometer (Mitutoyo Series 293) and the edema was calculated as the difference in thickness before treatment and 24, 48 and 72 h after challenge. The control group was treated only with oxazolone. Inhibition was expressed as a percentage of control. The mice were killed by means of cervical dislocation at 72 h and ear punches from each animal were used for determination of cytokine production. Determination of cytokine production in ear homogenates [16]: Ear samples were homogenized with a Polytron (Kinematica AG, Lucerne, Switzerland) in a buffer solution (10 mM HEPES pH = 7.6, 10 mM KCl, 1.5 mM MgCl2, 0.8% Triton X-100, 1 mM dithiotreitol, 2 mM phenylmethanesulfonyl fluoride, protease inhibitors cocktail, Roche), sonicated (3 x 10 s) and centrifuged at 14,000 rpm at 4ºC for 15 min. Supernatants were analyzed for protein content with Bradford reagent and frozen at –80ºC. IL-1β, IL-4 and TNF-α were quantified in homogenates in triplicate by a specific enzyme immunoassay kit used according to the manufacturer’s instructions (eBiosciences). After overnight incubation at 4ºC with the capture antibody, microplate wells were washed 3 times with washing buffer. Afterwards, the same operation was repeated after 1h incubation with diluent. Standards and test solutions (100 μL) were uploaded and incubated for 2 h at room temperature. Thereafter, Effects of terpenoids of Salvia willeana on hypersensitivity Natural Product Communications Vol. 3 (12) 2008 1957 two further steps consisting of the addition of biotin conjugate anti-mouse polyclonal antibody (100 μL, 1 h incubation) and avidin-peroxidase (100 μL, 30 min incubation) preceded the termination of the reaction with 1M H2SO4. Each incubation was followed by aspiration and repeated washing with the buffer. Final absorbance was read at 450 nm. Once a standard curve was obtained with different cytokine concentrations ranging from 7.81-1000 pg/mL, experimental values were calculated by means of interpolation. Inhibition percentage was defined as the difference between the mean value of control and test value absorbance, divided by control value, and multiplied by 100. obtained from the human blood of healthy volunteers. Cells were isolated by the Ficoll-Paque gradient density method (GE Healthcare). T lymphocytes were isolated by depletion of adherent cells on plastic dishes (95% purity) and resuspended in RPMI 1640 medium supplemented with 10% fetal bovine serum, 100 U/mL penicillin and 100 μg/mL streptomycin (Invitrogen Gibco, Langley, OK, USA). Determination of cell viability [17]: The cytotoxicity of the test compounds was performed by the 3-[4, 5dimethylthiazol-2-yl]-2,5-diphenyl-tetrazolium bromide (MTT) assay. Murine RAW 264.7 macrophages and human lymphocytes were exposed to test compounds at concentrations of 100, 50 and 25 μM in a microplate for various lengths of time and then 100 μL per well of a 0.5 mg/mL solution of MTT was added and incubated at 37°C until blue deposits were visible. The blue metabolite was dissolved in dimethyl sulfoxide (DMSO). Absorbance was measured at 570 nm using a Labsystems Multiskan EX plate reader (Midland, Canada). Results were expressed in absolute absorbance readings, with a decrease indicating a reduction in cell viability. A volume of 200 μL of T-lymphocyte suspension (1 x 106 cells/mL) was applied to each well of a 96-well plate with 25 μg/mL phytohemagglutinin (PHA) alone or with test compounds at different concentrations (5–10 μM) and dexamethasone (5 μM) as a positive control. The plates were incubated in a 5% CO2/air humidified atmosphere at 37°C for 3 days, after which T-cell proliferation was determined with a modified colorimetric MTT assay. The formazan product formed was dissolved in DMSO by shaking it. The absorbance was measured at 570 nm using a Labsystems Multiskan EX plate reader (Midland, Canada). Results were expressed in absolute absorbance readings; a decrease indicated a reduction in cell viability. Controls consisted either of lymphocytes with PHA (100% activity), with medium (0% activity) or samples with nonstimulated lymphocytes. Nitrite production in intact RAW 264.7 macrophages [18]: Murine RAW 264.7 macrophages were cultured in Dulbecco's Modified Eagle Medium (DMEM) containing 2 mM L-glutamine, 100 U/mL penicillin, 100 μg/mL streptomycin and 10% fetal bovine serum (all from Invitrogen). Cells were removed from the tissue culture flask with a cell scraper and resuspended to a final concentration of 1×106 cells/mL. Nitrite production was assessed as the index of nitric oxide generation in the induction phase. Thus, RAW 264.7 macrophages (1×106 cells/mL) were co-incubated in a 96-well culture plate (200 μL) with 1 μg/mL of lipopolysaccharide (Sigma-Aldrich) at 37° C for 24 h in the presence of test compounds (100, 50, 25 and 12.5 μM) or vehicle (phosphate-buffered saline). Nitrite concentration in culture supernatant was determined by using Griess reagent (Sigma-Aldrich). T-lymphocytes (1 x 106 cells/mL) were co-incubated in a 6-well plate with or without 25 μg/mL PHA after treatment with the compounds. Different experiments concerning the cell cycle were performed to see the effect on cell cycle after PHA stimulation and the effect of isolated compounds on PHA-stimulated cells at different times. Isolated compounds (6-8 μM) were added to the cells, after which the plates were incubated in a 5% CO2-air-humidified atmosphere at 37°C for 12, 48 and 72 h. In these experiments we see at what point the cell cycle is blocked and we view the subG0 peak as the index of the apoptosis event, and then the G1, G2 and S peaks. The cells were harvested by means of centrifugation, washed in phosphate-buffered saline (pH 7.2) and then fixed in 70% ethanol for 30 min at −20° C. After washing the cells once with phosphate-buffered saline, the DNA was stained with propidium iodide (4 μg/mL) containing 100 μg/mL of ribonuclease A. Flow cytometry analysis was conducted with an EPICS XCL (Beckman, Fullerton, CA, USA); 1 x 104 cells for each test sample were counted. T-lymphocyte isolation, proliferation and cell cycle analysis [19,20]: Peripheral lymphocytes were Statistics: Data were expressed as mean ± S.E.M. Statistical analysis involved carrying out a one-way 1958 Natural Product Communications Vol. 3 (12) 2008 analysis of variance (ANOVA) followed by Dunnett's t-test for multiple comparisons. In comparisons against the control group, values of P less than 0.05 were considered significant. Inhibition percentages (%I) were calculated from the differences between drug-treated groups and control animals treated only with the inflammatory agent. Vonaparti et al. Acknowledgments – This work was supported by the Spanish Ministry of Education and Science (FEDER) (SAF 2006-06726). The authors wish to thank Dr T. Vrahimi-Hadjilouca and Dr D. Droushiotis (Agricultural Research Institute, Ministry of Agriculture, Natural Resources & Environment, Nicosia, Cyprus) for providing us the plant material. References [1] Rustaiyan A, Masoudi S, Tabatabaei-Anaraki M. (2007) Terpenoids from Iranian Salvia species. Natural Product Communications, 2, 1031-1042. [2] Kintzios SE. (2000) The Genus Salvia. In Medicinal and aromatic plants: Industrial profiles Vol. 14, Gordon & Breach, Amsterdam. [3] Tsintidis T. (1995) The Endemic Plants of Cyprus. Editions of Bank of Cyprus Nicosia, Cyprus, 88. [4] Gennadios P. (1914) Dictionary of Medicinal Herbals. Leonis P. (Ed.). Athens, 300-301. [5] Ody P. (1993) The Herb Society’s Complete Medicinal Herbals. Dorling Kindesley Lt., London, 95. [6] De la Torre MC, Bruno M, Piozzi F, Savona G, Rodriguez B, Arnold NA. (1990) Terpenoids from Salvia willeana and S. virgata. Phytochemistry, 29, 668-670. [7] Huguet AI, Recio MC, Máñez S, Giner RM, Ríos JL. (2000) Effect of triterpenoids on the inflammation induced by protein kinase C activators, neuronally acting irritants and other agents. European Journal of Pharmacology, 410, 69-81. [8] Bani S, Kaul A, Khan B, Ahmad SF, Suri KA, Gupta BD, Satti NK, Qazi GN. (2006) Suppression of T lymphocyte activity by lupeol isolated from Crataeva religiosa. Phytotherapy Research, 20, 279-287. [9] Giner-Larza EM, Máñez S, Recio MC, Giner RM, Prieto JM, Cerdá-Nicolás M, Ríos JL. (2001) Oleanonic acid, a 3-oxotriterpene from Pistacia, inhibits leukotriene synthesis and has anti-inflammatory activity. European Journal of Pharmacology, 428, 137-143. [10] Raphael TJ, Kuttan G. (2003) Effect of naturally occurring triterpenoids glycyrrhizic acid, ursolic acid, oleanolic acid and nomilin on the immune system. Phytomedicine, 10, 483-489. [11] Raphael TJ, Kuttan G. (2003) Immunomodulatory activity of naturally occurring monoterpenes carvone, limonene, and perillic acid. Immunopharmacology and Immunotoxicology, 25, 285-294. [12] Friedrich K, Delgado IF, Santos LM, Paumgartten FJ. (2007) Assessment of sensitization potential of monoterpenes using the rat popliteal lymph node assay. Food of Chemistry and Toxicology, 45, 1516-1522. [13] Stevenso OE, Finch TM. (2003) Allergic contact dermatitis from rectified camphor oil in EarexR ear drops. Contact Dermatitis, 49, 42-52. [14] Dat NT, Lee IS, Cai XF, Shen G, Kim YH. (2004) Oleanane triterpenoids with inhibitory activity against NFAT transcription factor from Liquidambar formosana. Biological and Pharmacological Bulletin, 27, 426-428. [15] Topçu G. (2006) Bioactive triterpenoids from Salvia species. Journal of Natural Products, 69, 482-487. [16] Olmos A, Giner RM, Recio MC, Ríos JL, Cerdá-Nicolás JM, Máñez S. (2007) Effects of plant alkylphenols on cytokine production, tyrosine nitration and inflammatory damage in the efferent phase of contact hypersensitivity. British Journal of Pharmacology, 152, 374-385. [17] Mosmann T. (1983) Rapid colorimetric assay for the cellular growth and survival: application to proliferation and cytotoxicity assays. Journal of Immunology Methods, 65, 55-63. [18] Olmos A, Giner RM, Recio MC, Ríos JL, Máñez S. (2007) Modulation of protein tyrosine nitration and inflammatory mediators by isoprenylhydroquinone glucoside. European Journal of Pharmaceutical Sciences, 30, 220-228. [19] Kuo YC, Yang NS, Chou CJ, Lin LC, Tsai WJ. (2000) Regulation of cell proliferation, gene expression, production of cytokines, and cell cycle progression in primary human T-lymphocytes by piperlactam S isolated from Piper kadsura. Molecular Pharmacology, 58, 1057-1066. [20] Kuo YC, Weng SC, Chou CJ, Chang TT, Tsai WJ. (2003) Activation and proliferation signals in primary human T-lymphocytes inhibited by ergosterol peroxide isolated from Cordyceps cicadea. British Journal of Pharmacology, 140, 895-906. NPC Natural Product Communications Characterization of By-products of Saffron (Crocus sativus L.) Production 2008 Vol. 3 No. 12 1959 - 1962 Pamela Vignolinia, Daniela Heimlerb, Patrizia Pinellia, Francesca Ieria, Arturo Sciulloc and Annalisa Romani* a Dipartimento di Scienze Farmaceutiche, Università degli Studi di Firenze, via U. Schiff 6, 50019 Sesto Fiorentino, Italy b Dipartimento di Scienza del Suolo e Nutrizione della Pianta, Università degli Studi di Firenze, P.le delle Cascine 18, 50144 Firenze, Italy c ARPAT, Agenzia Regionale per la Protezione Ambientale della Toscana, via Ponte alle Mosse 211, 50144 Firenze, Italy annalisa.romani@unifi.it Received: July 28th, 2008; Accepted: October 31st, 2008 The stigma, stamens and sepals of Crocus sativus L,. from two different geographical origins, were analyzed for their crocin and flavonol contents. Identification of crocins, safranal, picrocrocin, and flavonols was carried out by HPLC/DAD and HPLC/MS analysis. Both stigma samples, grown under natural conditions, exhibited high crocin contents (between 342 and 231 mg/g), while the stamens and sepals were rich in flavonols (between 6 and 10 mg/g). The stamens contain mainly kaempferol- 3-O-sophoroside, whereas the sepals contain mainly quercetin and methyl-quercetin glycosides. These data may be useful in order to find a possible exploitation of the by-products of saffron production, in which large quantities of C. sativus flowers are available. Keywords: Crocins, flavonols, HPLC/DAD/MS, sepals, stamens, stigma. The dried, red stigmas of Crocus sativus L. are a very expensive spice known as saffron, which is used as a food flavoring and coloring agent and as a traditional herbal medicine [1a]. Crocus is cultivated in India, Iran, Spain, Greece and Italy. The production process involves a large amount of manual work and cannot be completely mechanized. In Italy, from a 1000 m2 area, about 120,000-150,000 flowers can be obtained (4000-5000 kg), which give rise to 5-7 kg of fresh stigma, i.e. 1.0-1.3 kg of dried product. Many papers deal with methods for the separation and determination of the biologically active [1b-1f] and aroma components [2a-2c]. The quality control of commercial saffron is checked using spectrophotometric [3a,3b], TLC [3c], GC [3d], HPLC [3e], and CE [3f] methods. The purpose of this paper is the analysis of stigmas from C. sativus cultivated in Italy (Perugia and Fiesole) in order to characterize this commercial saffron from a quality point of view. In these areas, cultivation is effected under natural conditions and without the use of any chemical product in the drying and conservation phases. However, the most important part deals with the characterization of the biologically active components of the stamens and sepals in order to find a possible use for this material, which forms the major part of C. sativus flowers. The exploitation of stamens and sepals, notwithstanding their availability as by-products in the production of saffron, has not been taken into account, with the exception of one paper dealing with the isolation of flavonoids from crocus petals to study their tyrosinase inhibition action [4a]. Notwithstanding the lack of information on the polyphenol content of these tissues, petal extracts were used to control rat blood pressure [4b] and to test their antitussive effect in guinea pigs [1b]. The major biologically active components of saffron are crocin analogues, which are all glycosides of 1960 Natural Product Communications Vol. 3 (12) 2008 O-R2 CH3 CH3 O Table 1: Quantitative data for dried stigma. Average value ± SD of three samples. Data are expressed as mg/g fresh sample. O O-R1 CH3 COMPOUNDS (Rt) CH3 COMPOUND R1 R2 MW Crocin-5 ; C50H74O29 Crocin-4 ; C44H64O24 Crocin-3 ; C38H54O19 Crocin-2 ; C33H44O14 Crocin-2’ ; C33H44O14 Crocin-1 ; C26H34O9 Crocetin ; C20H24O4 Three β-glucosyl β-D-gentiobiosyl 1138 β-D-gentiobiosyl β-D-gentiobiosyl 976 β-D-gentiobiosyl β-D-glucosyl 814 β-D-gentiobiosyl H 652 β-D-glucosyl β-D-glucosyl 652 β-D-glucosyl β-D-glucosyl 490 H H 328 CHO CHO HO Gluc.-O Safranal C10H14O MW= 150 Picrocrocin C16H26O7 MW= 330 OH HO O R OH OH Vignolini et al. COMPOUND R MW Quercetin ; C15H10O7 ΟΗ 302 Kaempferol ; C15H10O6 Η 286 O Figure 1: Chemical structures of saffron components trans-crocetin, a carotenoid derivative, and which are responsible for the color. Safranal (2,6,6-trimethyl1,3-cyclohexadien-1-carboxaldehyde), which is responsible for the characteristic aroma of saffron, is formed during storage by dehydration of picocrocin, which is responsible for its bitter taste. Flavonoids are found in stigma, sepals, and stamens (Figure 1). As regards stigma, the composition of the extract was similar to that found by other authors regarding crocins, picocrocins, and safranal. Three kaempferol derivatives (two triglycosides and one diglycoside) were identified, according to previous findings [1f,5a]. In the case of stamens, a lesser number of crocins was found and quercetin, as well as kaempferol derivatives were detected. Also, methylquercetin derivatives in quite large amounts were recorded. There were no differences, from a qualitative point of view, between the two sampling zones; in fact only a quantitative variation was found in the samples from the different geographic regions [5a]. Table 1 reports the quantitative data for the dried stigma. It should be noted that the two samples differ are present in largest amount in the two samples. These compounds, together with cis-crocin 4, were trans crocin-5 (10.30) crocin derivative (11.14) crocin derivative (11.46) crocin derivative (11.87) trans crocin-4 (12.84) crocin derivative (13.88) trans crocin-3 (14.39) crocin derivative (14.99) crocin derivative (15.90) trans crocin-2' (16.17) crocin derivative (17.37) cis crocin-4 (17.79) trans crocin-2 (19.33) crocin derivative (20.40) crocin derivative (21.11) cis crocin-1 (22.02) crocin derivative (22.81) crocin derivative (23.17) TOTAL Picrocrocin (6.34) Safranal (24.87) K-3-sophoriside -7- glucoside (3.78) K -3,7,4'-triglucoside (5.90) K-3-sophoroside (8.49) TOTAL Stigma (FI) Stigma (PG) 2.4±0.09 2.1±0.08 0.3±0.01 0.3±0.009 238.9±2.86 1.3±0.06 65.6±1.84 0.2±0.01 0.6±0.03 2.1±0.07 0.3±0.01 9.5±0.33 16.9±0.51 0.2±0.009 0.3±0.01 1.0±0.05 0.2±0.01 traces 342.02 111.1±2.33 2.2±0.09 4.7±0.2 1.2±0.05 6.2±0.22 2.0±0.07 0.8±0.04 0.3±0.01 0.1±0.007 148.5±2.66 0.5±0.02 46.2±1.38 0.2±0.009 0.5±0.02 1.5±0.06 0.3±0.009 14.1±0.49 14.8±0.50 traces traces 0.8±0.04 traces 0.5±0.02 231.1 68.9±1.79 2.6±0.09 3.3±0.14 0.9±0.04 5.4±0.17 12.1 9.64 mainly in trans-crocin 4, trans-crocin 3 and picrocrocin contents, i.e. the three compounds which also the main compounds found by Caballero-Ortega et al. [5b] in a study of 11 saffron samples from different origins. The crocins content of the two samples is quite high giving evidence for the very good quality of the two samples. Among flavonols, kaempferol-3-O-sophoroside was the main compound reported for a Spanish sample analyzed by Carmona et al. [5a]. Table 2 reports the crocin contents of sepals and stamens. The amount of crocins is low, while that of flavonols (Table 3) ranged from 10.1 to 6.1 mg/g. Stamens and sepals differ mainly in their kaempferol3-O-sophoroside content, which is the most abundant flavonol in the sepals. The flavonols composition of the two tissues is different: in sepals, kaempferol derivatives ranged between 91 -93 %, whereas in stamens, quercetin and methyl-quercetin derivatives ranged between 5271%. From all these data the possible exploitation of alternative tissues like stamens and sepals as phytochemical resources can be pointed out. For each kg of stigma, about 1000 kg of flowers are processed; therefore, sepals and stamens are important by-products of saffron production and their use could increase the economic value of C. sativus flowers. Characterization of byproducts of saffron Natural Product Communications Vol. 3 (12) 2008 1961 Table 2: Crocins content of sepals and stamens. Average value ± SD of three samples. COMPOUNDS (Rt) trans crocin-4 (12.84) crocin der. (13.88) trans crocin-3 (14.39) crocin der. (14.99) crocin der. (15.99) trans crocin-2' (16.17) cis crocin-4 (17.79) trans crocin-2 (19.33) cis crocin-1 (22.02) crocin der. (22.81) crocin der. (23.17) cis crocin-2 (24.82) Sepals (FI) Sepals (PG) Stamens (FI) Stamens (PG) 3.1±0.17 traces 112.2±5.65 4.0±0.19 1.7±0.09 traces 33.4±1.74 traces traces traces 0.8±0.04 traces 1.3±0.07 traces traces traces 3.3±0.18 traces 22.0±1.14 0.1±0.006 20.7±1.07 1.3±0.08 7.0±0.38 traces 0.3±0.02 traces 0.1±0.008 traces 0.3±0.02 TOTAL 4.2 0.6±0.03 traces 196.3 5.4 Experimental Sample preparation: Sepals, stamens and dried stigma samples were obtained from plants harvested in 2005 from Fiesole (FI, Italy) and Perugia (PG, Italy). Sepals and stamens (500 mg) were suspended in 50 mL of 70% ethanol, adjusted to pH 2.0 with formic acid, and left overnight. After extraction, the samples were filtered to eliminate plant residues, and the filtrate evaporated to dryness under vacuum at room temperature. The residue was redissolved in EtOH/H2O (70:30) and adjusted to pH 2.0 with formic acid to a final volume of 3 mL. Saffron stigmas (50 mg) were extracted with 10 mL of 70% ethanol, adjusted to pH 2.0 with formic acid, left overnight and then filtered to eliminate plant residues. The extracts were analysed by HPLC/DAD/MS for the determination of saffron components. Authentic standards of crocin were purchased from Fluka (St. Louis, USA), safranal from Sigma-Aldrich (St. Louis, USA), and p-hydroxybenzoic acid, kaempferol 3-O-glucoside, rutin and curcumin from Extrasynthèse S.A. (Lyon, France). All solvents were of HPLC grade purity (BDH Laboratory Supplies, United Kingdom). HPLC/DAD analysis:. Analysis for flavonols and crocins was carried out using a HP 1100L liquid chromatograph equipped with a DAD detector and managed by a HP 9000 workstation (Agilent Technologies, Palo Alto, CA, USA). Flavonols and crocins were separated by using a 150 × 3.9 mm i.d. 4 μm Nova-Pak C18 column (Waters) operating at 27°C. UV/Vis spectra were recorded in the 190-600 nm range and the chromatograms were acquired at 250, 308, 350 and 440 nm. The mobile phase was a Table 3: Flavonols content of sepals and stamens. Average value ± SD of three samples. Data are expressed as μg/g fresh sample. COMPOUNDS (Rt) Kaempferol derivative (3.71) Kaempferol-3-sophoroside-7-glucoside (3.78) Kaempferol derivative (5.81) Kaempferol diglucoside (5.89) Kaempferol derivative (6.49) Kaempferol diglucoside (7.30) Quercetin diglucoside (7.30) Methyl quercetin diglucoside (7.82) Quercetin derivative (8.15) Methyl quercetin di glucoside (8.42) Kaempferol-3-sophoroside (8.49) Kaempferol glucosyl rhamnoside (9.29) Methyl quercetin derivative (9.34) Quercetin derivative (9.44) Quercetin diglucoside (9.58) Kaempferol sinapoyl glucoside (10.59) Kaempferol derivative (10.86) Kaempferol glucoside (10.98) Methyl quercetin glucoside (11.13) Quercetin derivatives (11.55-12.21) Kaempferol derivative (12.99) Quercetin p-cumaroyl glucoside (13.76) Quercetin derivative (14.09) Kaempferol p-cumaroyl glucoside (15.42) Methyl quercetin p-cumaroyl glucoside (15.61) Kaempferol (18.43) TOTAL Sepals (FI) 76±4.10 Sepals (PG) 97±4.85 15±1.03 113±5.6 34±1.83 480±22.08 82±4.16 738±32.16 84±4.21 6415±192.45 41±2.13 8304±215.9 66±3.20 24±1.27 306±14.38 60±3.18 309±14.25 421±19.78 399±18.75 Stamen(FI) Stamen(PG) 511±22.84 24±1.18 923±41.53 77±4.15 416±21.16 1037±37.32 628±28.88 27±1.15 209±10.03 1702±64.7 755±33.75 1227±47.81 2091±61.74 39±2.14 249±11.73 377±17.72 691±31.09 239±11.47 1188±46.32 303±14.54 140±5.81 39±2.25 21±1.15 93±4.65 52±2.75 26±1.19 17±0.078 199±9.75 4±0.22 35±2.05 26±1.21 20±1.16 7998 176±8.62 66±3.43 14±0.74 10138 6059 237±110.61 26±1.20 52±2.65 40±2.12 8±0.44 7873 1962 Natural Product Communications Vol. 3 (12) 2008 one-step linear solvent gradient system, starting from 90% H2O (adjusted to pH 3.2 with HCOOH) up to 100% CH3CN during a 60-min period; flow rate 0.8 mL min-1. HPLC/MS analysis: HPLC/MS analysis was performed using a HP 1100L liquid chromatograph linked to a HP 1100 MSD mass spectrometer with an API/electrospray interface (Agilent Technologies, Palo Alto, CA, USA). The mass spectrometer operating conditions were: gas temperature, 350°C; nitrogen flow rate, 10.5 L/min, nebulizer pressure, 40 psi; quadrupole temperature, 30°C; and capillary voltage, 3500 V. The mass spectrometer was operated in positive mode at 120 eV. Identification and quantification of individual polyphenols: Quantification of individual compounds was directly performed by HPLC/DAD using a five-point regression curve (r2 ≥ 0.998) in the range Vignolini et al. 0-30 μg on the basis of authentic standards. In particular, crocin derivatives were determined at 440 nm using curcumin as reference compound; safranal was determined at 308 nm using safranal as reference compound and picrocrocin was determined at 250 nm using p-hydroxybenzoic acid as reference compound. Flavonols, like kaempferol and quercetin derivatives, were determined at 350 nm using kaempferol-3-Oglucoside and rutin, respectively, as reference compounds. In all cases, actual concentrations of the derivatives were calculated after applying corrections for differences in molecular weight. Acknowledgments - The authors wish to express their sincere gratitude to the Cassa di Risparmio di Firenze that contributed to the acquisition of a part of the instrumentation used for this work. We express sincere thanks to Mr Piscolla, Azienda Agricola Poggio al Sole (Fiesole, FI) for the supply of saffron samples from Fiesole. References [1] [2] [3] [4] [5] (a) Xi L, Qian Z. (2006) Pharmacological poperties of crocetin and crocin (digentiobiosyl) ester of crocetin from. saffron. Natural Product Communications, 1, 65-75; (b) Alonso GL, Salinas M., Garijo J, Sanchez-Fernandez MA. (2001) Composition of crocins and picocrocin from Spanish saffron (Crocus sativus L.), Journal of Food Quality, 24, 219-233; (c) Li N, Lin G, Kwan YW, Min ZD. (1999) Simultaneous quantification of five major biologically active ingredients of saffron by high-performance liquid chromatography. Journal of Chromatography A, 849, 349-355; (d) Pfander H, Rychener M. (1982) Separation of crocetin glycosyl esters by high-performance liquid chromatography. Journal of Chromatography A, 234, 443-447; (e) Tarantilis PA, Polissiou M, Manfait M. (1994) Separation of picrocrocin, cis-trans-crocins and safranal of saffron using high-performance liquid chromatography with photodiode-array detection. Journal of Chromatography A, 664, 55-61; (f) Tarantilis PA, Tsoupras G, Polissiou M. (1995) Determination of saffron (Crocus sativus L.) components in crude plant extract using high-performance liquid chromatography-UV-visible photodiode-array detection-mass spectrometry. Journal of Chromatography A, 699, 107-118 (a) Lozano P, Delgado D, Gomez D, Rubio M, Iborra JL. (2000) A non-destructive method to determine the safranal content of saffron (Crocus sativus L.) by supercritical carbon dioxide extraction combined with high performance liquid chromatography and gas chromatography. Journal of Biochemical and Biophysical Methods, 43, 367-378; (b) Loskutov AV, Beninger CW, Hosfield GL, Sink KC. (2000) Development of an improved procedure for extraction and quantitation of safranal in stigmas of Crocus sativus L. using high performance liquid chromatography. Food Chemistry, 69, 87-95; (c) Straubinger M, Bau B, Eckstein S, Fink M, Winterhalter P. (1998) Identification of novel glycosidic aroma precursors in saffron (Crocus sativus L.). Journal of Agricultural and Food Chemistry, 46, 3238-3243. (a) Carmona M, Carrion ME, Zalacain A, Alonso GL. (2004) Detection of adulterated saffron through UV-Vis spectral analysis. Journal of Food Science & Technology, 41, 451-455; (b) Zalacain A, Ordoudi SA, Blazquez I, Diaz-Plaza EM, Carmona M, Tsimidou MZ, Alonso GL. (2005) Screening method for the detection of artificial colours in saffron using derivative UV-Vis spectrometry after precipitation of crocetin. Food Additives and Contaminants, 22, 607-615; (c) Corti P, Mazzei E, Ferri S, Granchi GG, Dreassi E. (1996) High performance thin layer chromatographic quantitative analysis of picrocrocin and crocetin, active principles of saffron (Crocus sativus L.): a new method. Phytochemical Analysis, 7, 201-203; (d) Alonso GL, Salinas MR, Garijo J. (1998) Method to determine the authenticity of aroma of saffron (Crocus sativus L.). Journal of Food Protection, 61, 1525-1528; (e) Lozano P, Castellar MR, Simancas MJ, Iborra JL. (1999) Quantitative high-performance liquid chromatographic method to analyse commercial saffron (Crocus sativus L.) products. Journal of Chromatography A, 830, 477-483; (f) Zougagh M, Simonet BM, Rios A, Valcarcel M. (2005) Use of non-aqueous capillary electrophoresis for the quality control of commercial saffron samples. Journal of Chromatography A, 1085, 293-298. (a) Kubo I, Kinst-Hori I. (1999) Flavonols from saffron flower: tyrosinase inhibitory activity and inhibition mechanism. Journal of Agricultural and Food Chemistry, 47, 4121-4125; (b) Fatehi M, Rashidabady T, Fatehi-Hassanabad Z. (2003) Effects of Crocus sativus petals extract on rat blood pressure and on responses induced by electrical field stimulation in the rat isolated vas deferens and guinea pig ileum. Journal of Ethnopharmacology, 84, 199-203; (c) Hosseinzadeh H, Ghenaati J. (2006) Evaluation of the antitussive effect of stigma and petals of saffron (Crocus sativus) and its components safranal and crocin in guinea pig. Fitoterapia, 66, 446-448. (a) Carmona M, Sanchez AM, Ferreres F, Zalacain A, Tomas-Barberan F, Alonso GL. (2007) Identification of the flavonoids fraction in saffron spice by LC/DAD/MS/MS: comparative study of samples from different geographic origins. Food Chemistry, 100, 445-450; (b) Caballero-Ortega H, Pereda-Miranda R., Abdullaev FI. (2007) HPLC quantification of major active components from 11 different saffron (Crocus sativus L.) sources. Food Chemistry, 100, 1126-1131. NPC Natural Product Communications Antitrypanosomal and Antileishmanial Activities of Organic and Aqueous Extracts of Artemisia annua 2008 Vol. 3 No. 12 1963 - 1966 Anna Rita Biliaa, Marcel Kaiserb, Franco Francesco Vincieria and Deniz Tasdemirc,* a Department of Pharmaceutical Sciences, University of Florence, 50019 Sesto Fiorentino, Florence, Italy b Department of Medical Parasitology, Swiss Tropical Institute, 4002 Basel, Switzerland c Centre for Pharmacognosy and Phytotherapy, School of Pharmacy, University of London, London WC1N 1AX, UK deniz.tasdemir@pharmacy.ac.uk Received: July 22nd, 2008; Accepted: October 17th, 2008 Artemisia annua is an herbal drug with profound antimalarial activity, which can be ascribed to the sesquiterpene lactone artemisinin. Artemisinin also shows efficacy against other parasitic protozoan species, such as Trypanosoma and Leishmania, however trypanocidal and leishmanicidal effects of A. annua extracts have not been reported so far. In the current study, we evaluated the in vitro growth inhibitory activity of a number of organic and aqueous A. annua extracts, including tinctures, infusions and decoctions against three parasitic protozoa, T. brucei rhodesiense, T. cruzi and L. donovani. Artemisinin content of these extracts was determined by HPLC/DAD/MS. Artemisinin was also evaluated for its antiparasitic activity for comparison. Among the tested extracts, the acetone- and the n-hexane-solubles of A. annua were the most potent against T. b. rhodesiense with IC50 values of 0.30 and 0.455 μg/mL, respectively, whereas the other extracts were ten- to fifty-fold less potent. Neither of the extracts nor artemisinin had trypanocidal activity against T. cruzi (IC50 > 30 μg/mL). Only the organic extracts of A. annua arrested the growth of L. donovani with modest IC50 values (5.1 to 9.0 μg/mL) comparable to that of artemisinin (IC50 8.8 μg/mL). This study highlights significant variations in the artemisinin content of A. annua extracts and underlines the potential of A. annua extracts and artemisinin in the treatment of trypanosomal and leishmanial infections. Keywords: Artemisia annua, organic and aqueous extracts, artemisinin, Trypanosoma, Leishmania. Artemisia annua L. (Asteraceae) is an annual herb that has been used against fever in traditional Chinese medicine for over 2,000 years [1a]. The plant is nowadays listed in the Pharmacopoeia of the Republic of China for the treatment of fever and malaria. The recommended daily dose is specified as 4.5 to 9 g of dried herb prepared as a tea (infusion) with boiling water [1b]. After the discovery of artemisinin (1) as the antimalarial principle of the plant, all clinical studies have focused on this compound. Currently artemisinin-based combination therapy (ACT) is the most effective treatment of drug-resistant malaria and recommended by the WHO. A. annua is the sole source of artemisinin, which occurs in the plant in very low concentrations, resulting in high costs of therapy. Thus, there is an increasing interest in the use of A. annua teas in malaria endemic areas where large scale pharmaceutical production is not possible. Accordingly, the consumption of A. annua tea or decoctions by patients in clinical studies has been shown to reduce parasitaemia significantly [1c]. Trypanosomiasis and leishmaniasis are insect-borne parasitic diseases that represent a significant and neglected public health problem worldwide. Trypanosoma brucei rhodesiense is the causative agent of African trypanosomiasis (sleeping sickness) in South and East Africa, infecting 50,000 people every year, primarily in the poorest rural populations of sub-Saharan Africa where the tsetse fly vector is common [1d]. The incidence of the disease may approach 500,000 cases per year. The American trypanosomiasis, also known as Chagas’ disease caused by T. cruzi is a major endemic disease in South and Central America. It is estimated that 18 million people are infected with the disease and 50,000 individuals die of it annually [2]. Leishmaniasis is a tropical and subtropical disease affecting nearly 88 countries, with an estimated global prevalence of 12 million cases annually. It is caused by protozoal parasites of the genus Leishmania and occurs in three different clinical forms (cutaneous, muco-cutaneous and visceral), where visceral leishmaniasis (Kala azar), due to 1964 Natural Product Communications Vol. 3 (12) 2008 L. donovani and L. infantum, is the most severe form. Leishmaniasis presents as an opportunistic infection in association with immunodepression, in particular AIDS [3a]. Both trypanosomiasis and visceral leishmaniasis are invariably fatal if untreated. There is no prophylactic chemotherapy and little or no prospect of a vaccine. Moreover, current treatments of these diseases are far from being ideal because of the high costs, long treatment courses, high clinical failure and serious toxic, even fatal side effects. Thus, it is obvious that new drugs and therapy regimes are needed for the treatment of these diseases. Besides its profound antimalarial effect, artemisinin has been shown to possess growth inhibitory effects against pathogenic Trypanosoma and Leishmania species [3b,3c]. However, the effects of different A. annua extracts have not been studied. In a previous study on a selected high-yield Brazilian cultivar of A. annua, we determined the artemisinin content of a number of solvent extracts, such as aqueous EtOH (tinctures AA-T40, AA-T60), infusions (AA-I1, AA-I2, AA-I3) and decoctions (AA-D1, AA-D2) by HPLC/DAD/MS and compared these with the artemisinin concentration in the n-hexane extract (AA-Hexane) [3d]. In the continuation of our studies on this A. annua cultivar, we have now prepared additional solvent extracts, namely toluene (AA-Toluene), dichloromethane (AA-DCM), acetone (AA-Acetone) and 100% ethanol (AA-EtOH). Their artemisinin content was determined by HPLC/DAD/MS, as previously reported [3d,3e]. All available extracts, as well as artemisinin were tested for in vitro activity against T. brucei rhodesiense, T. cruzi and L. donovani. Table 1 displays the quantitative analysis results on artemisinin content of all twelve extracts investigated. Percentages are given on the dried extracts obtained by evaporation of the organic solvent or freeze-dried aqueous solutions, and do not reflect the content of artemisinin in the dried herbal drug, which is about 0.52%. Very different artemisinin percentages were detected in the investigated extracts, with AA-DCM being the richest with 3.68% artemisinin. This extract was followed by EtOH, n-hexane and acetone extracts (2.64%, 2.28% and 1.82, respectively), whereas the toluene extract was the poorest (0.57%). The tinctures prepared with 40% v/v (AA-T40) and 60% v/v (AA-T60) EtOH had also lower artemisinin yields (0.75% and 1.08%), similar to those of A. annua infusions or decoctions, which contained roughly 0.7-0.8% artemisinin. Bilia et al. Table 1: Artemisinin content, trypanocidal and leishmanicidal activities of different solvent extracts of A. annua and artemisinin (IC50 values are in μg/mL and mean values from at least two replicates, i.e. the variation is a maximum of 20%). Extract/ Compound AA-Toluene AA-Acetone AA-Hexane AA-DCM AA-EtOH AA-T40 AA-T60 AA-I1 AA-I2 AA-I3 AA-D1 AA-D2 Artemisinin Reference Extraction yield (g) 3.01 2.47 2.28 0.68 1.04 1.80a 1.90a 2.24a 2.10a 2.78a 2.36a 2.31a Artemisinin (%±SEM) 0.57±0.07 1.82±0.11 2.28±0.98 3.68±1.01 2.64±0.93 0.75±0.16a 1.08±0.20a 0.72±0.11a 0.68±0.23a 0.80±0.14 a 0.68±0.13a 0.81±0.15 a T. b. rhodesiense 3.31 0.30 0.455 3.82 14.57 13.31 23.48 21.15 22.15 22.58 22.96 24.40 24.40 0.004a T. cruzi >30 >30 >30 >30 >30 >30 >30 >30 >30 >30 >30 >30 >30 0.22b L. donovani 9.0 5.10 5.64 8.50 >30 >30 >30 >30 >30 >30 >30 >30 8.80 0.10c a : These results were obtained in our previous study [3d] Reference compounds: amelarsoprol, bbenznidazole, cmiltefosine. The extracts with most abundant artemisinin levels are shown bold. Table 1 also displays the IC50 values of the extracts investigated and artemisinin against T. brucei rhodesiense, T. cruzi and L. donovani. The AAAcetone and AA-Hexane extracts showed the highest trypanocidal effect against African trypanosome (T. b. rhodesiense) with IC50 values of 0.30 and 0.455 μg/mL. The next best activity was exerted by AAToluene and AA-DCM, which had almost ten times higher IC50 values (3.31 μg/mL and 3.82 μg/mL, respectively). AA-T40 and AA-EtOH extracts showed comparable but reduced trypanocidal effects (IC50 values 13.31 and 14.57 μg/mL, respectively), whereas all remaining extracts exhibited IC50 values above 20 μg/mL. The potency of artemisinin was similar (IC50 24.4 μg/mL). It is noteworthy that the trypanocidal effect of acetone and n-hexane extracts is much greater than that of artemisinin. This finding may be due to the presence of other bioactive compounds in these extracts or compounds having significant synergistic effect with artemisinin. On the other hand, none of the extracts or artemisinin appeared to have the ability to inhibit the growth of American trypanosome, T. cruzi at the highest concentrations tested (IC50 values >30 μg/mL). Of the twelve extracts that were studied for in vitro antileishmanial activity, only the organic A. annua extracts proved to have reasonable potential. Notably AA-Acetone and AA-Hexane extracts had almost identical activities (IC50 values 5.1 and 5.64 μg/mL), as did the AA-DCM and AA-Toluene extracts (IC50 values 8.5 and 9.0 μg/mL). All other remaining extracts, including AA-EtOH were inactive (IC50 >30 μg/mL). Leishmanicidal activity of artemisinin was also comparable (IC50 8.80 μg/mL) to AA-DCM. Trypanocidal and leishmanicidal activities of A. annua extracts Natural Product Communications Vol. 3 (12) 2008 1965 To our knowledge, this is the first time that A. annua extracts have been studied for trypanocidal and leishmanicidal properties. Although the results might appear low in comparison to standard drugs, organic A. annua extracts, in particular the acetone and nhexane extracts still represent good activity against T. b. rhodesiense and L. donovani. These two extracts merit further investigations for the identification of their active components, which might be different from artemisinin. boiling water, then left to cool and filtered through a filter paper. Sample I2: A 9 g sample of dried aerial parts of A. annua was extracted with 1 L of boiling water, covered, then left to cool and filtered through a filter paper. Sample I3: A 9 g sample of dried aerial parts of A. annua was extracted with 1 L of boiling water, then left to cool for 15 min and filtered through a filter paper. For analytical purposes, all infusions (filtrates) were freeze-dried and directly analysed. Experimental HPLC-DAD and HPLC-MS systems: The HPLC analyses were performed using a HP 1100 Liquid Chromatograph (Agilent Technologies, Palo Alto, CA, USA) equipped with a HP 1040 Diode Array Detector (DAD), an automatic injector, an auto sampler, a column oven and managed by a HP 9000 workstation (Agilent Technologies, Palo Alto, CA, USA). Separations were performed on a reversed phase column (Purospher®Star RP-18, namely Hibar®). The HPLC system was interfaced with a HP 1100 MSD API-electrospray (Agilent Technologies, Palo Alto, CA, USA). The interface geometry, with an orthogonal position of the nebulizer with respect to the capillary inlet, allowed the use of analytical conditions similar to those of HPLC-DAD analysis. Mass spectrometry operating conditions were optimized in order to achieve maximum sensitivity values: gas temperature 350°C at a flow rate of 10 L/min, nebulizer pressure 30 p.s.i., quadrupole temperature 30°C, and capillary voltage 3500 V. Full scan spectra from m/z 100 to 800 in the positive ion mode were obtained (scan time 1 s). A prepacked column RT (250 x 4.6 mm) with particle size 5 µm (Merck, Darmstadt, Germany) was employed. The eluents were A: water adjusted to pH 3.2 with formic acid; B: acetonitrile; C: methanol. The system was operated with the oven temperature at 26oC. Before HPLC analysis, each sample was filtered through a cartridge-type sample filtration unit with a polytetrafluoroethylene (PTFE) membrane (d=13 mm, porosity 0.45 µm, (Lida Manufacturing Corp.) and immediately injected. Chromatograms were recorded between 200 and 450 nm. DAD spectra were stored for all peaks exceeding a threshold of 0.1 mAu. Plant material: The selected high-yield cultivar of A. annua [3f] was provided by P.M. Magalhães of the Universidade Estadual de Campinas (Brazil). Dried aerial parts of the plant were used in all analyses. Chemicals: Artemisinin was purchased from Sigma (Sigma-Aldrich). All the solvents used for the extraction and HPLC analysis (EtOH, toluene, MeOH, n-hexane, dichloromethane, and acetone) were HPLC grade from Merck (Darmstadt, Germany); 85% formic acid was provided by Carlo Erba (Milan, Italy). Water was purified by a MilliQplus system from Millipore (Milford, MA). Preparation of organic extracts: Small pieces of dried aerial parts of A. annua (10 g) were exhaustively extracted at room temperature by maceration with 100 mL of organic solvent (toluene, n-hexane, acetone, dichloromethane or EtOH) for 72 h, separately. The filtrates were subsequently evaporated to dryness under reduced pressure to obtain the crude organic extract. Preparation of the tinctures: Small pieces of dried aerial parts of A. annua (10 g) were extracted at room temperature by maceration with 100 g of ethanol [either 40 or 60% v/v (samples T40 and T60)]. The solutions were concentrated under vacuum and freeze-dried prior to the analysis. Preparation of the decoctions: Sample D1: A 9 g sample of dried aerial parts of A. annua was extracted with 1 L of boiling water, kept boiling for 5 min, then left to cool and filtered through a filter paper. Sample D2: A 9 g sample of dried aerial parts of A. annua was extracted with 1 L of boiling water, kept boiling for 5 min and immediately filtered through a filter paper. For analytical purposes, both D1 and D2 (filtrates) were freeze-dried and directly analysed. Preparation of the infusions: Sample I1: A 9 g sample of dried A. annua was extracted with 1 L of Calibration curves: A calibration curve, obtained from a methanolic solution of artemisinin (1 mg/mL), was used to quantify artemisinin in the extracts and tinctures, while a methanolic solution of artemisinin (0.5 mg/mL) was employed to determine the artemisinin content of infusions and decoctions. 1966 Natural Product Communications Vol. 3 (12) 2008 Sample analysis: Samples of 5 mg of the different extracts were accurately weighed and suspended in methanol (1.0 mL). The suspensions were sonicated for 10 min and filtered through a cartridge-type sample filtration unit before HPLC analysis. The tinctures were injected as prepared. In vitro assay for Trypanosoma brucei rhodesiense: Minimum Essential Medium (50 μL) supplemented according to [4a] with 2-mercaptoethanol and 15% heat-inactivated horse serum was added to each well of a 96-well microtiter plate. Serial drug dilutions were prepared covering a range from 90 to 0.123 μg/mL. Then 104 bloodstream forms of Trypanosoma b. rhodesiense STIB 900 in 50 μL were added to each well and the plate incubated at 37°C under a 5% CO2 atmosphere for 72 h. After addition of 10 μL of Alamar Blue™ to each well, the plates were incubated for another 2-4 h and read in a Spectramax Gemini XS microplate fluorometer using an excitation wavelength of 536 nm and emission wavelength of 588 nm. Fluorescence development was expressed as percentage of the control, and IC50 values determined. In vitro assay for Trypanosoma cruzi: Rat skeletal myoblasts (L6 cells) were seeded in 96-well microtiter plates at 2000 cells/well in 100 μL RPMI 1640 medium with 10% FBS and 2 mM L-glutamine. After 24 h, the medium was removed and replaced by 100 μL per well containing 5000 trypomastigote forms of T. cruzi Tulahuen strain C2C4 containing the β-galactosidase (Lac Z) gene. After 48 h, the medium was removed from the wells and replaced by 100 μL fresh medium with or without a serial drug Bilia et al. dilution. After 96 h of incubation, the substrate CPRG/Nonidet (50 μL) was added to all wells. A color reaction, developed within 2-6 h, was read photometrically at 540 nm. Data were transferred into a graphic program (MS Excel™); sigmoidal inhibition curves were determined and IC50 values calculated. In vitro assay for Leishmania donovani: Amastigotes of Leishmania donovani strain MHOM/ET/67/L82 were grown in axenic culture at 37°C in SM medium [4b] at pH 5.4 supplemented with 10% heat-inactivated fetal bovine serum under an atmosphere of 5% C02 in air. Culture medium (100 μL) with 1 x 105 amastigotes from axenic culture with or without a serial drug dilution were seeded in 96-well microtiter plates. Seven 3-fold dilutions were used covering a range from 30 to 0.041 μg/mL. After 72 h of incubation, 10 μL of Alamar Blue™ was added to each well. The plates were incubated for another 2 h and read with a Spectramax Gemini XS microplate fluorometer (Molecular Devices Cooperation, Sunnyvale, CA, USA) using an excitation wavelength of 536 nm and an emission wavelength of 588 nm. Data were analyzed using the software Softmax Pro (Molecular Devices Cooperation, Sunnyvale, CA, USA). Decrease of fluorescence (= inhibition) was expressed as percentage of the fluorescence of control cultures and plotted against the drug concentrations. From the sigmoidal inhibition curves the IC50 values were calculated. Acknowledgments – Authors thanks Prof. P.M. Magalhães of the Universidade Estadual de Campinas (Brazil) for providing the plant material. References [1] [2] [3] [4] (a) van Agtmael MA, Eggelte TA, van Boxtel CJ. (1999) Artemisinin drugs in the treatment of malaria: from medicinal herb to registered medication. Trends in Pharmacological Sciences, 20, 199-205.; (b) Stöger EA. (1991) Arzneibuch der Chinesischen Medizin. Deutscher Apotheker-Verlag, Stuttgart, 37-45; (c) Räth K, Taxis K, Walz G, Gleiter CH, Li SM, Heide L. (2004) Pharmacokinetic study of artemisinin after oral intake of a traditional preparation of Artemisia annua L. (annual wormwood). American Journal of Tropical Medicine and Hygiene, 70, 128-32; (d) The World Health Report (2004) Changing History (World Health Organisation, Geneva). Fact Sheet Number 116; World Health Organization: Geneva, Switzerland. World Health Organization 2003. Chagas’ disease, http://www.who.int/tdr/ dw/chagas2003.htm (a) Berhe N, Wolday D, Hailu A, Abraham Y, Ali A, Gebre-Michael T, Desjeux P, Sonnerborg A, Akuffo H, Britton S. (1999) HIV viral load and response to antileishmanial chemotherapy in co-infected patients. AIDS, 13, 1921-1925; (b) Mishina YV, Krishna S, Haynes RK, Meade J.C. (2007) Artemisinins inhibit Trypanosoma cruzi and Trypanosoma brucei rhodesiense in vitro growth. Antimicrobial Agents and Chemotherapy, 51, 1852-1854; (c) Sen R, Bandyopadhyay S, Dutta A, Mandal G, Ganguly S, Saha P, Chatterjee M. (2007) Artemisinin triggers induction of cell-cycle arrest and apoptosis in Leishmania donovani promastigotes. Journal of Medical Microbiology, 56, 1213-1218; (d) Bilia AR, Gabriele C, Bergonzi MC, Melillo de Malgalhaes P, Vincieri FF. (2006) Variation in artemisinin and flavonoid content in differerent extracts of Artemisia annua. Natural Product Communications, 1, 1111-1115; (e) Bilia AR, Melillo de Malgalhaes P, Bergonzi MC, Vincieri FF. (2006) Simultaneous analysis of artemisinin and flavonoids of several extracts of Artemisia annua L. obtained from a commercial sample and a selected cultivar. Phytomedicine, 13, 487-493; (f) Magalhães PM, Delabays N, Sartoratto A. (1997) Ciência e Cultura. Journal of Brazilian Association for the Advancement of Science, 49, 413-415. (a) Baltz T, Baltz D, Giroud C, Crockett J. (1985) Cultivation in a semi-defined medium of animal infective forms of Trypanosoma brucei, T. equiperdum, T. evansi, T. rhodesiense and T. gambiense. EMBO Journal, 4, 1273-1277; (b) Cunningham I. (1977) New culture medium for maintenance of tsetse tissues and growth of trypanosomatids. Journal of Protozoology, 24, 325-329. NPC 2008 Vol. 3 No. 12 1967 - 1970 Natural Product Communications Secondary Metabolites from the Roots of Salvia palaestina Bentham Antonio Vassalloa, Ammar Baderb, Alessandra Bracac, Angela Bisiod, Luca Rastrellia, Francesco De Simonea and Nunziatina De Tommasia,* a Dipartimento di Scienze Farmaceutiche, Università di Salerno, Via Ponte Don Melillo, 84084 Fisciano (SA), Italy b Faculty of Pharmacy, Al-Zaytoonah Private University of Jordan, P.O. Box 130, 11733 Amman, Jordan c Dipartimento di Chimica Bioorganica e Biofarmacia, Università di Pisa, Via Bonanno 33, 56126 Pisa, Italy d Dipartimento di Chimica e Tecnologie Farmaceutiche ed Alimentari, Università di Genova, Via Brigata Salerno 13, 16147 Genova, Italy detommasi@unisa.it Received: July 25th, 2008; Accepted: November 3rd, 2008 Two new sesquiterpenes (1-2), and one diterpene (3) were isolated from the roots of Salvia palaestina Bentham (Lamiaceae), together with eight known diterpenes and two triterpenes. Their structures were elucidated by 1D and 2D NMR spectroscopy, including 1D-TOCSY, DQF-COSY, ROESY, HSQC, and HMBC experiments, as well as ESIMS and chemical analysis. Keywords: Salvia palaestina, Lamiaceae, sesquiterpenes, diterpenes, NMR. The genus Salvia L. belongs to the Lamiaceae family and is represented by numerous species, widely distributed in various regions of the world, used for many medicinal and pharmaceutical applications, including as a rich source of essential oils [1a,1b]. S. palaestina Bentham, known also as Palestine sage, is a perennial herb, 30-70 cm long, lemon scented during the flowering season, distinguishable by the basal leaves, which are pinnatilobed or pinnatisect, oblong or lanceolate; the upper lip of the corolla is large, exceeding 10 mm and the corolla has pure white or rarely purplish colour, while the floral leaves are membranous, with white or pink colour [1c]. The plant is widespread in the Eastern Mediterranean area and in the Western Irano-Turanian regions. In Turkish folk medicine a preparation made from an extract of the leaves is commonly used as a wound healer [2a]. Previous phytochemical studies on the aerial parts of the plant reported the presence of modified abietane diterpenoids, sesquiterpenes, diterpenes, sesterterpenes, triterpenes, and flavonoids [2a-2d]. As part of a continuing investigation of Salvia species [3a-3d], we made a phytochemical study of S. palaestina roots collected in Jordan and herein we 14 H 2 10 H 9 1 3 5 4 8 6 H H3COCO O 7 12 H O 11 15 13 1 2 OH O HOOC 3 Figure 1: Structures of compounds 1-3 report the structural characterization of three new terpenoids (1-3, Figure 1) from the apolar extract of the title plant, on the basis of extensive spectroscopic and spectrometric analysis, including 2D NMR and ESIMS spectra. Eight known diterpenes and two triterpenes were also isolated and characterized as ferruginol [4a], 6β-hydroxyferruginol [4b], pisiferic acid [4c], dehydroabietane-11,12-diol [4d], carnosic acid [4e], 12-O-demethylcryptojaponol [4f], 12-deoxy-6-hydroxy-6,7-dehydroroyleanone [4g], aethiopinone 1968 Natural Product Communications Vol. 3 (12) 2008 Table 1: 1H NMR spectroscopic data of compounds 1 and 2 (CD3OD, 600 MHz)a. position 1 2a 2b 3a 3b 5 6 7 8a 8b 9a 9b 12 13 14a 14b 15 COCH3 3’ 4’ 5’ Vassallo et al. Table 2: 13C NMR spectroscopic data of compounds 1 and 2 (CD3OD, 150 MHz)a. compounds 1 2 δH δH 2.22 ddd (10.0, 9.6, 8.8) 2.25 ddd (10.0, 9.6, 8.8) 1.96 m 1.98 m 1.58b 1.58b 1.78 ddd (10.0, 3.5, 2.0) 1.80 ddd (10.0, 3.5, 2.0) 1.58b 1.61b 1.41 dd (11.5, 9.5) 1.44 dd (11.3, 9.5) 0.53 dd (11.5, 9.5) 0.52 dd (11.3, 9.5) 0.72 ddd (11.5, 9.5, 6.0) 0.70 ddd (12.0, 9.5, 6.0) 2.00 m 2.00 m 1.06 m 1.08 m 2.43 ddd (14.0, 7.2, 1.0) 2.40 ddd (14.0, 7.0, 1.0) 2.07 m 2.07 m 1.04 s 1.04 s 1.04 s 1.04 s 4.70 br s 4.70 br s 4.66 br s 4.66 br s 1.25 s 1.27 s 1.99 s 6.10 br q (6.5) 2.00 d (6.5) 1.87 s δ values were established from the 1D-TOCSY, DQF-COSY, HSQC and HMBC experiments.b overlapped signals. position 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 COCH3 COCH3 1’ 2’ 3’ 4’ 5’ a [4h], lupeol [2c], and ursolic acid [2c], respectively, by comparison of their spectroscopic data with those reported in the literature. Compound 1 was isolated as an amorphous powder and yielded a pseudo-molecular ion in the positive HR ESIMS at m/z 263.1912. The NMR (Tables 1 and 2) and mass spectral data of 1 indicated it to have the molecular formula C17H26O2, thus having five elements of unsaturation. Analysis of its 13C NMR spectrum showed the presence of one acetyl group (carbonyl signal at δC 170.0 and methyl signal at δC 21.0 and δH 1.99) and one 1,1-substituted C-C double bond (δC 153.5 and 105.0) as the only multiple bonds within the molecule, permitting the recognition of 1 as a tricyclic sesquiterpene. Identifiable from 13C NMR spectroscopic data was also a resonance consistent with one carbinolic carbon (δC 84.0). The 1 H NMR spectrum of 1 exhibited two methyl singlets and one double intensity singlet at δ 1.04, 1.04, 1.25, and 1.99 and a cyclopropyl moiety [δ 0.53 (1H, dd, J = 11.5 and 9.5 Hz) and δ 0.72 (1H, ddd, J = 11.5, 9.5, and 6.0 Hz)]. Analysis of 1D-TOCSY and DQF-COSY experiments allowed the sequential assignments of hydrogens from H-1 to H-3 and from H-5 to H-9. HSQC and HMBC experiments provided unambiguous assignments of all the proton and carbon resonances. This information, together with the HMBC spectral data, finally led to the assignment of an aromadendrane-like skeleton for 1 [5a]. HMBC compounds 1 δC 54.0 26.2 41.4 84.0 53.8 30.0 27.1 24.4 39.8 153.5 20.4 28.0 28.0 105.0 23.6 170.0 21.0 2 δC 54.2 26.3 41.5 82.0 54.0 31.1 27.0 24.5 39.8 153.0 20.5 28.2 28.2 105.5 23.8 169.0 127.7 139.0 15.6 20.6 δ values were established from the 1D-TOCSY,DQF-COSY, HSQC and HMBC experiments. a correlations between H-3a⎯C-5, H-5⎯C-4, Me-15⎯C-3, H-6⎯C-4 Me-15⎯C-5 substantiated the presence of an acetoxy group at C-4. The relative stereochemistry of 1 was ascertained by ROESY experiments. By irradiation of the signal for H-1 [δ 2.22 (1H, ddd, J = 10.0, 9.6, 8.8 Hz)], a ROE effect with the signal of H-6 was observed, indicating them to be on the same side of the molecule and on the opposite side to H-5. Further ROEs were detected between H-5 and Me-13, H-6 and H-7, and H-7 and Me-12. Therefore, compound 1 was determined as 4-O-acetylspathulenol. The HR ESIMS of 2 gave a pseudo molecular ion at m/z 303.2285 (C20H30O2). The NMR data of 2 (Tables 1 and 2) were very similar to those of 1, suggesting the same skeleton. The NMR spectra revealed the absence of the acetyl group in 2 replaced by a 2-methyl-2-butenoyl moiety [δ 6.10 (1H, br q, J = 6.5 Hz), δ 2.00 (3H, d, J = 6.5 Hz); δ 1.87 (3H, s)]. The stereochemistry of 2 was supported by NMR experiments and deduced to be the same as compound 1. Consequently, the new compound 2 was characterized as 4-O-(2-methyl-2butenoyl)spathulenol. The molecular formula of 3 was determined as C20H34O4 by the HR ESIMS ion at m/z 339.2438 [M+H]+, as well as from its 13C and 13C DEPT NMR spectroscopic data. Terpenoids from Salvia palaestina Natural Product Communications Vol. 3 (12) 2008 1969 Table 3: 1H (600 MHz) and 13C NMR (150 MHz)spectroscopic data of compound 3 (CD3OD)a. position 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 3 δH 1.69b 0.98 1.66b 1.50b 1.80b 1.50b 1.84 dd (11.0, 4.2) 1.67b 1.70 ddd (12.0, 10.0, 4.0) 1.46b 1.20 dd (12.0, 3.2) 1.44b 1.32b 1.90b 1.52b 1.68b 1.52b 3.60 d (4.0) 3.54 d (4.0) 1.26 s 1.31 s 1.15 s 0.79 s δC 37.8 18.6 38.2 48.2 52.4 20.8 42.7 75.0 56.9 37.0 21.8 29.2 75.0 44.7 62.0 26.4 25.4 185.0 16.0 15.0 δ values were established from 1D-TOCSY, DQF-COSY, HSQC and HMBC experiments. b overlapped signals. a Comparison of NMR data (Table 3) of 3 with those of 13-epi-manoyloxide-18-oic acid indicated that 3 is an epi-manoyl oxide derivative [5b]. In particular, protons and carbons due to rings A-C resonated at almost the same frequencies as the corresponding signals in 13-epi-manoyloxide-18-oic acid, while the side chain NMR signals were the point of difference. Particularly, NMR spectral data revealed the absence of the signals for the –CH=CH2 group in 3 in respect to those of 13-epi-manoyloxide-18-oic acid and the presence of signals for a –CH2CH2OH group. These data, together with mass spectral analysis were consistent with 3 being 15-hydroxy-8,13epoxylabdan-18-oic acid. A literature survey indicates that the aerial parts of Salvia species contain flavonoids, triterpenoids, and monoterpenes, particularly in the flowers and leaves, while diterpenoids are found mostly in the roots. Some American Salvia contained also diterpenoids in the aerial parts, and in a few Salvia species, triterpenoids and flavones are present in the roots [5c]. The present investigation of S. palaestina roots is completely in agreement with our results obtained from the aerial parts of the plant [2d]. Sesquiterpenes with an aromadendrane-like skeleton, diterpenes, sesterterpenes, and triterpenes are characteristic constituents of the plant; also hypogeal organs lacked sesterterpenes. Experimental General experimental procedures: Optical rotations were measured on a Perkin-Elmer 241 polarimeter equipped with a sodium lamp (589 nm) and a 1 dm microcell. All the NMR spectra were acquired in CD3OD in the phase-sensitive mode with the transmitter set at the solvent resonance and TPPI (Time Proportional Phase Increment) used to achieve frequency discrimination in the ω1 dimension. The standard pulse sequence and phase cycling were used for all 2D experiments. The NMR data were processed on a Silicon Graphic Indigo2 Workstation using UXNMR software. Column chromatography was performed on silica gel (63-200 μm, Merck, Darmstadt, Germany); high resolution mass spectra were acquired on a Q-Tof Premier instrument (Waters, Milford, MA), equipped with a nanospray ion source; to achieve high accuracy mass measurements, both external and internal calibrations of the spectrometer were performed using quercetin (molecular mass 302.0427) or amentoflavone (molecular mass 538.0900) as standards. HPLC separations were conducted on a Waters 590 system equipped with a Waters R401 refractive index detector, and with a Waters μ-Bondapak C18 column (Waters, Milford, MA). TLC was performed on precoated Kieselgel 60 F254 plates (Merck, Darmstadt, Germany); compounds were detected with Ce(SO4)2/H2SO4 solution. Plant material: The whole plant of Salvia palaestina Bentham was collected in April 2005 in Amman, Jordan, and identified by Dr Ammar Bader. A voucher specimen, number Jo-It-2003/2, has been deposited in the Herbarium of the Laboratory of Pharmacognosy and Phytochemistry at Al-Zaytoonah Private University of Jordan. The roots were separated from the aerial parts; the latter were investigated previously [3a]. Extraction and isolation: The dried roots of S. palaestina (290 g) were finely powdered and exhaustively extracted by maceration at room temperature with acetone to give 3.0 g of residue. This was subjected to column chromatography using silica gel and eluting with n-hexane followed by increasing concentrations of CHCl3 (between 50% and 100%), and by increasing concentrations of MeOH in CHCl3 (between 1% and 100%). Fractions 1970 Natural Product Communications Vol. 3 (12) 2008 of 40 mL were collected, analysed by TLC (silica gel plates, in CHCl3-n-hexane 1:1, CHCl3 or mixtures CHCl3-MeOH 99:1, 98:2, 97:3, 9:1, 4:1), and grouped into 13 fractions. Fraction 2 contained pure aethiopinone (30 mg). Fraction 3 (76 mg) was rechromatographed over silica gel eluting with n-hexane followed by increasing concentration of CHCl3 (between 1% and 100%) to obtain pure pisiferic acid (7.5 mg) and 12-deoxy-6-hydroxy-6,7dehydroroyleanone (20 mg). Fraction 4 (573 mg) was subjected to silica gel column chromatography, eluting with n-hexane followed by increasing concentration of CHCl3 (between 50% and 100%), to give six major fractions (A-F). Fractions B (40 mg), C (80 mg), and D (35 mg) were separately subjected to RP-HPLC on a C18 μ-Bondapak column (30 cm x 7.8 mm, flow rate 2.0 mL min-1) with MeOH-H2O (4:1) to yield pure 6β-hydroxyferruginol (10 mg, tR = 16 min) from fraction B, carnosic acid (2 mg, tR = 22 min), compound 1 (2 mg, tR = 24 min), and compound 2 (5 mg, tR = 30 min), from fraction C, and dehydroabietane-11,12-diol (4 mg, tR = 16 min), carnosic acid (2.5 mg, tR = 22 min), and 12-Odemethylcryptojaponol (3 mg, tR = 23 min) from fraction D, respectively. Fractions 7 (32 mg) and 9 Vassallo et al. (52 mg) were separately chromatographed over RPHPLC on a C18 μ-Bondapak column (30 cm x 7.8 mm, flow rate 2.0 mL min-1) with MeOH-H2O (68:32) to give pure compound 3 (6 mg, tR = 12 min) and ferruginol (2 mg, tR = 14 min) from fraction 7, and ursolic acid (4 mg, tR = 34 min) and lupeol (3 mg, tR = 38 min) from fraction 9, respectively. Compound 1 [α]D: +23 (c 1.00, MeOH). 1 H and 13C NMR: Table 1 and Table 2. HR ESIMS: m/z 263.1912 [M+H]+, calcd for C17H26O2 262.1933. Compound 2 [α]D: +27 (c 1.00, MeOH). 1 H and 13C NMR: Table 1 and Table 2. HR ESIMS: m/z 303.2285 [M+H]+ calcd for C20H30O2 302.2246. Compound 3 [α]D: +25 (c 1.00, MeOH). 1 H NMR and 13C NMR: Table 3. HR ESIMS: m/z 339.2438 [M+H]+ calcd for C20H34O4 338.2457. References [1] [2] [3] [4] [5] (a) Foster S, Tyler VE. (2000) Tyler’s Honest Herbal. The Haworth Press, Binghamton, New York, 327-330; (b) Steinegger E, Hansel R. (1988) Lehrbuch der Pharmakognosie. Springer Verlag: Berlin; (c) Al-Eisawi D. (1998) Field Guide to Wild Flowers of Jordan and neighbouring countries. Jordan Press Foundation Al-Rai, Amman, 175. (a) Miski M, Ulubelen A, Johansson C, Mabry TJ. (1983) Antibacterial activity studies of flavonoids Salvia palaestina. Journal of Natural Products, 46, 874-875; (b) Ulubelen A, Miski M, Johansson C, Lee E, Mabry TJ, Matlin SA. (1985) Terpenoids from Salvia palaestina. Phytochemistry, 24, 1386-1387; (c) Hussein AA, de La Torre MC, Rodriguez B, Hammouda FM, Hussiney HA. (1997) Modified abietane diterpenoids and a methoxylupane derivative from Salvia palaestina. Phytochemistry, 45, 1663-1668; (d) Hussein AA, Rodriguez B. (2000) A diterpenoid with a new modified abietane skeleton from Salvia palaestina. Zeitschrift fuer Naturforschung, B: Chemical Sciences, 55, 233-234. (a) Cioffi G, Bader A, Malafronte A, Dal Piaz F, De Tommasi N. (2008) Secondary metabolites from the aerial parts of Salvia palaestina. Phytochemistry, 69, 1005-1012; (b) Malafronte A, Dal Piaz F, Cioffi G, Braca A, Leone A, De Tommasi N. (2008) Secondary metabolites from the aerial parts of Salvia aethiopis L. Natural Product Communications 3, 877-880; (c) De Felice A, Bader A, Leone A, Sosa S, Della Loggia R, Tubaro A, De Tommasi N. (2006) New polyhydroxylated triterpenes and antiinflammatory activity of Salvia hierosolymitana. Planta Medica, 72, 643-649; (d) Bisio A, De Tommasi N, Romussi G. (2004) Diterpenoids from Salvia wagneriana. Planta Medica, 70, 452-457. (a) Connolly JD, Hill RA. (1991) Dictionary of Terpenoids, vol. 2 Di- and higher terpenoids. Chapman & Hall: London; (b) Kuo YH, Yu MT. (1996) Diterpenes from the heartwood of Juniperus formosana Hay. var. concolor Hay. Chemical & Pharmaceutical Bulletin, 44, 1431-1435; (c) Lin TC; Fang JM, Cheng YS. (1999) Terpenes and lignans from leaves of Chamaecyparis formosensis. Phytochemistry, 51, 793-801; (d) Sanchez AJ, Konopelski JP. (1994) The first synthesis of (±)-taxodone. Synlett 5, 335-336 (e) Bruno M, Savona G, Piozzi F, De la Torre MC, Rodriguez B, Marlier M. (1991) Abietane diterpenoids from Lepechinia meyeni and Lepechinia hastata. Phytochemistry, 30, 2339-2343; (f) Rodriguez B. (2003) 1H and 13C NMR spectral assignments of some natural abietane diterpenoids. Magnetic Resonance in Chemistry, 41, 741-746; (g) Nagy G, Gunther G, Mathe I, Blunden G, Yang MH, Crabb TA. (1999) 12-Deoxy-6,7-dehydroroyleanone, 12-deoxy-6-hydroxy-6,7-dehydroroyleanone and 12-deoxy-7,7-dimethoxy-6ketoroyleanone from Salvia nutans roots. Phytochemitsry, 51, 809-812; (h) Lin LZ, Blasko G, Cordell GA (1989) Diterpenes of Salvia prionitis. Phytochemistry, 28, 177-181. (a) Inagaki F, Abe A. (1985) Analysis of proton and carbon-13 nuclear magnetic resonance spectra of spathulenol by twodimensional methods. Journal of the Chemical Society, Perkin Transactions 2: Physical Organic Chemistry, 1773-1778; (b) Zetadero C, Bohlmann F, King RM. (1992) Ent-labdanes manoyloxide and elipterol derivatives from Chrysocephalum ambiguum. Phytochemistry, 31, 1631-1638; (c) Bozan B, Ozturk N, Kosar M, Tunalier Z, Baser KHC. (2002) Antioxidant and free radical scavenging activities of eight Salvia species. Chemistry of Natural Compounds (Translation of Khimiya Prirodnykh Soedinenii), 38, 198-200. NPC Natural Product Communications Cancer Chemopreventive Potential of Humulones and Isohumulones (Hops α- and Iso-α-acids): Induction of NAD(P)H:Quinone Reductase as a Novel Mechanism 2008 Vol. 3 No. 12 1971 - 1976 Gregor Bohra, Karin Klimob, Josef Zappa, Hans Beckera and Clarissa Gerhäuserb,* a Fr. 8.2 Pharmakognosie und Analytische Phytochemie der Universität des Saarlandes, Saarbrücken, Germany b Toxikologie und Krebsrisikofaktoren, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany c.gerhauser@dkfz.de Received: June 10th, 2008; Accepted: August 31st, 2008 Phytochemical analysis and chemopreventive testing of a special “α-/β-acid free” hops extract led to the identification of isohumulones (hops iso-α-acids) as potent inducers of NAD(P)H:quinone reductase (QR) activity. CD values (concentrations required to double the specific activity of QR in Hepa1c1c7 cell culture) were in the range of 1.3 to 10.2 µg/mL, with CD value of trans-isohumulone < cis-isoadhumulone < cis-isocohumulone < cis-isohumulone (+ trans-isoadhumulone). Humulones (hops α-acids) were equally active with CD values of 3.4 to 7.6 µg/mL. However, these activities were accompanied by cytotoxicity. Cohumulinone and humulinone, oxidation products of co- and n-humulone, were inactive. We further identified isohumulones as potent inhibitors of lipopolysaccharide-induced inducible nitric oxide synthase (iNOS) activity in Raw264.7 cell culture, with IC50 values of 5.9 – 18.4 µg/mL. Humulones and humulinones were inactive at concentrations < 20 µg/mL. These results indicate that isohumulones, which are considered as the most abundant class of polyphenols in beer, should by further investigated for chemopreventive efficacy in animal models. Keywords: hops, Humulus lupulus L., cancer chemoprevention, NAD(P)H:quinone reductase, hops α-acids, hops iso-α-acids, humulone, isohumulone, humulinone. Hops (Humulus lupulus L.) have been used since ancient times for brewing [1]. It was soon realized that they not only added bitterness and aroma to beer, but also played an important role as a preservative. Subsequently, hops α- and β-acids (humulones and lupulones), constituents of the essential bitter resin, were identified as strong antibiotics against Grampositive bacteria ([2] and literature cited therein). β-Acids are extremely sensitive to oxidation and do not survive the brewing process. During wort-boiling, the poorly water-soluble humulones are isomerized to isohumulones (iso-α-acids), which are better soluble; this process is involved in the generation of the bitter flavor of beer [3a]. Isohumulones also play an important role in foam stabilization [3b]. Overall, they represent one of the most abundant classes of polyphenols in beer; concentrations of up to 100 mg/L have been reported in very bitter English ales [3c]. Isohumulones are optically active molecules which occur as cis- and trans-isomers. In analogy to the chemical structures of humulones, three isoforms indicated by the prefix “co-“, “-n-“ and “ad-“ are present in beer, which differ only in their acyl side chain (summary in Figure 1). Interestingly, Hughes reported that cis-isohumulones were more bitter than their trans-isomers. In particular, bitterness of the compounds was described as cis-isohumulone > trans-isohumulone ≈ cis-isocohumolone > transisocohumulone [3b]. In recent years, hops have gained considerable interest because of the biological and potential cancer chemopreventive activities of some of their constituents (reviewed in [4a-c]). As an example, the α-acid n-humulone was described as a potent antioxidant and anti-inflammatory agent capable of inhibiting the induction of cyclooxygenase-2 (Cox-2) in cell culture and mouse skin [5], and displayed 1972 Natural Product Communications Vol. 3 (12) 2008 O OH R= Co- R O HO R= O n- HO O R= Humulones (α-acids) Ad- R= Acyl-side chain of humulones and isohumulones O H H O R HO OH O cis-Isohumulones (cis-iso-α-acids) OH HO O O O OH Cohumulinone R HO OH O trans-Isohumulones (trans-iso-α-acids) OH HO O O O OH n-Humulinone Figure 1: Chemical structures of humulones, cis- and transisohumulones, co- and n-humulinone. anti-proliferative activity by induction of cell differentiation and apoptosis in HL-60 human promyelocytic leukemia cells in vitro. It also inhibited angiogenesis in the chick embryo chorioallantoic membrane (CAM) assay, with a halfmaximal effective concentration ED50 of 1.5 µg/CAM. Topical application of n-humulone (1 mg) very potently suppressed tumor incidence induced by 12-O-tetradecanoylphorbol-13-acetate (TPA) in the two-stage mouse skin model by 93% and tumor multiplicity by 99%. In addition to these cancer preventive effects, humulone was described as a very potent inhibitor of bone resorption and a candidate therapeutic agent for osteoporosis, with a half-maximal inhibitory concentration of 5.9 nM (2.1 ng/mL) in an in vitro model of pit formation. Cohumulone was inactive at a concentration of 1 µM (reviewed in [4c]). Little information is available regarding potential cancer chemopreventive activities of isohumulones. Nozawa et al. demonstrated that freeze-dried beer at a dose of 1%, and isomerized hops extract (IHE) at 0.01 or 0.05% in the diet significantly reduced azoxymethane-induced preneoplastic precursor lesions in rat colon. IHE also potently reduced levels of prostaglandin E2 (PGE2) in colonic mucosa, Bohr et al. indicating anti-inflammatory potential by inhibition of Cox-2 expression [6a]. Several reports have suggested that isohumulones may have beneficial effects for the treatment of diabetic symptoms by inhibition of aldose reductase and reduction of insulin resistance, hyperlipidemia and obesity by activation of peroxisome proliferator-activated receptor (PPAR) α and γ [6b-f]. They were also shown recently to reduce renal injury in salt-sensitive rats by antioxidant activity [6g]. In continuation of our studies on hops prenylflavonoids [7] and acylphloroglucinol derivatives [8], we here describe results of the phytochemical analysis and chemopreventive testing of a special hops extract which led to the separation of four isohumulones and humulinone, an oxidation product of n-humulone. The chemopreventive potential of these compounds was compared with that of the α-acids cohumulone, n-humulone and adhumulone. For the isolation of isohumulones we fractionated a commercially available “α-/β-acid free” ethanolic hops extract [9] by size exclusion column chromatography into 18 fractions. Bitter-tasting fraction X08 was separated by semi-preparative HPLC to yield five subfractions X08.A to X08.E. Comparison of NMR and ESI mass spectra with those published [10a-f] led to the following peak assignment: Peak 1 was identified as “n-humulinone”, peak 2 as “cis-isocohumulone”, peak 3 as “trans-isohumulone”, peak 4 as “cis-isohumulone” (maybe plus “trans-isoadhumulone), and peak 5 as “cis-isoadhumulone”. For comparison of potential cancer chemopreventive activities, co-, n-, and adhumulone were isolated from a hops CO2-extract by counter-current chromatography (modified from [11]). Co- and n-humulinone were synthesized starting from co- and n-humulone according to [10b]. Identities were confirmed by comparison with published spectral data (see Experimental). Cancer chemoprevention has been defined as the use of chemical agents, natural products or dietary components to block, inhibit, or reverse the development of cancer in normal tissue and preneoplastic lesions [12]. Carcinogenesis is a multi-stage process, which is generally divided into initiation, promotion and progression phases and could be regarded as a continuous accumulation of biochemical and genetic cell damage. The cascade of QR induction by humulones and isohumulones Natural Product Communications Vol. 3 (12) 2008 1973 Table 1: Summary of potential chemopreventive activities. Fractions/ Compds X08 X08.A X08.B X08.C X08.D X08.E n-Humulinone Cohumulinone n-Humulone Cohumulone Adhumulone DPPHa SC50 >200 132.3 75.2 112.5 74.9 95.5 >200 >200 5.0 7.2 11.9 QR CD 2.6 5.0 7.0 1.3 10.2 5.6 >20 >20 3.4 6.7 7.6 IC50 >20 >20 >20 >20 >20 >20 >20 >20 4.0 9.4 11.5 iNOS IC50 >20 18.1 18.4 5.9 >20 9.8 >20 >20 >20 >20 >20 a Test systems: DPPH: DPPH scavenging (SC50 in µg/mL); QR: QR induction (CD, concentration required to double the specific activity of QR in µg/mL, IC50 for toxicity in µg/mL); iNOS: iNOS inhibition (IC50 in µg/mL). events resulting in tumor formation offers a variety of targets for intervention at every stage. Accordingly, fraction X08, its five subfractions X08.A – X08.E containing isohumulones, as well as the purified humulones and humulinones were tested in a series of test systems indicative of cancer chemopreventive potential. Manifestation of oxidative stress by infections, immune diseases and chronic inflammation has been associated with carcinogenesis in the initiation and promotion phase. Antioxidants may prevent the formation of highly reactive oxidation products, activation of carcinogens, formation of oxidized DNA bases and DNA strand breaks, which have been associated with overproduction of reactive oxygen species (ROS) and are involved in the carcinogenic process [13]. We determined radical scavenging potential by reaction with 1,1-diphenyl-2picrylhydrazyl (DPPH) free radicals. Consistent with an earlier report [14], the α-acids n-, co- and adhumulone were identified as potent radical scavengers with half-maximal scavenging concentrations (SC50) of 5.0 µg/mL (13.7 µM), 7.2 µg/mL (20.6 µM) and 11.9 µg/mL (32.9 µM), respectively, as indicated in Table 1. These activities were attributed to the presence of a hydroxyl-group in position C-5 [14] and were only slightly lower than those of ascorbic acid (SC50: 8.5 µM) and the watersoluble Vit. E analog Trolox (SC50: 9.7 µM) [15]. In contrast, fractions containing isohumulones were about 10-fold less potent in scavenging DPPH radicals than the humulones, with SC50 values in the range of 75 – 132.3 µg/mL. Oxidation to n-humulinone and cohumulinone further reduced antioxidant activities. Both compounds scavenged DPPH radicals less than 50% at the maximum test concentration of 200 µg/mL. Xenobiotics, including carcinogens, are metabolized and generally detoxified during phase 1 and 2 metabolism. Phase 2 enzymes, such as glutathione Stransferases (GST), conjugate phase 1 metabolites with endogenous ligands and thus enhance their excretion in the form of these conjugates. NAD(P)H:quinone reductase (QR) is not a conjugating enzyme. However, it contributes to detoxification of reactive quinones by 2-electron reduction, thereby preventing the formation of reactive semiquinones and ROS formation by redox cycling [16]. QR activity is induced coordinately with other phase 2 enzymes, making it a well established marker for potential chemopreventive activity [15]. Using QR induction in murine Hepa1c1c7 cell culture as a test system, we identified isohumulones as very potent inducers of QR activity (Table 1). All fractions dose-dependently induced QR activity in a concentration range of 1.25 to 20 µg/mL (Figure 2). Fraction X08.C containing trans-isohumulone was identified as the most active fraction followed by fractions X08.A and X08.E. Humulones also demonstrated good QR-inducing potential with CD values (concentration required to double QR activity) in the range of 3.4 to 7.6 µg/mL. In contrast to isohumulones, these compounds were toxic to the utilized murine hepatoma cells with IC50 values of 4.0 to 11.5 µg/mL. The ratio between IC50 values and CD values, previously defined as Chemopreventive Index, was close to 1, indicating that these compounds may stimulate their own detoxification [15]. Oxidation of humulones to humulinones completely abrogated QR-inducing potential, but also cytotoxicity. Induction of QR activity by humulones and isohumulones may be explained by activation of the transcription factor Nrf2/Keap-1 pathway similar to other natural products containing “Michael acceptor” functionality [17]. Chronic infections and inflammation lead to nuclear factor κB (NF-κB)-dependent induction of proinflammatory enzymes, such as Cox-2 and inducible nitric oxide synthase (iNOS). (Over)production of NO has been linked to early steps in carcinogenesis via nitrosative desamination of DNA bases, accumulation of reactive nitrogen oxide species and DNA adduct formation [18]. We and others have shown previously that induction of QR activity is often related to inhibition of iNOS induction [19a-c]. It was, therefore, of interest to analyze whether humulones and isohumulones would inhibit iNOS induction, using the murine macrophage cell line Raw264.7 stimulated with bacterial lipopolysaccharides (LPS) as a model. 1974 Natural Product Communications Vol. 3 (12) 2008 Bohr et al. QR induction (T/C) 6 4 1.25 µg/mL 2.5 µg/mL 5.0 µg/mL 10.0 µg/mL 20.0 µg/mL 2 0 X08 X08.A X08.B X08.C X08.D X08.E n- coadhumulone Figure 2: Induction of NAD(P)H:quinone reductase (QR) activity in Hepa1c1c7 cell culture. QR induction was computed by comparison with a solvent treated control (T: treated/ C: control). In correlation with QR induction, fraction X08.C was most potent in inhibiting LPS-induced iNOS activity with an IC50 value of 5.9 µg/mL. These data were in agreement with previous findings of Nozawa et al., who reported that isomerized hops extract and isohumulone inhibited PGE2 production by Cox-2 in LPS/interferon-γ-stimulated Raw264.7 macrophages [6a]. Neither humulones nor humulinones inhibited iNOS induction in our test system at concentrations up to 20 µg/mL (Table 1). In contrast, TNF-αmediated Cox-2 expression was potently inhibited by humulone in the murine osteoblastic MC3T3-E1 cell line [20]. Mechanistic investigations indicated that transcription factors NF-κB and NF-IL6 were targets of humulone action. The observed discrepancy of results obtained with humulone in the MC3T3-E1 and Raw264.7 cell lines may be due to differences in the utilized inducers and variations in the signal transduction machinery in both cell lines. Humulone was also reported to inhibit Cox-2 enzymatic activity with an IC50 of 1.6 µM [20]. We were not able to reproduce this result at concentrations up to 5 µM using human recombinant Cox-2 as an enzyme source (data not shown, method as described in [7]). In addition to these studies on humulone and isohumulones, Zhao et al. have investigated the potential of other hops constituents, including the β-acid lupulone, xanthohumol, and a series of derivatives of both compounds, to inhibit NO production in the Raw macrophage system [21]. Only chalcones such as xanthohumol were identified as potent inhibitors in this study, whereas lupulone and the β-acid derivatives were inactive. In conclusion, the present report provides first evidence that induction of phase 2 metabolizing enzymes could contribute to humulone- and isohumulone-mediated cancer chemoprevention. We have identified humulones and isohumulones as novel potent inducers of QR activity. Taking into consideration the relatively high concentrations of isohumulones in beer compared with other bioactive hops components, such as xanthohumol [7], further investigations on potential cancer chemopreventive efficacy and their influence on phase 2 metabolizing enzymes in animal models are warranted. A first indication may be seen in the dose-dependent reduction of carcinogen-induced mammary carcinogenesis by freeze-dried beer [22]. In that study, Nozawa et al. also demonstrated that feeding rats with freeze-dried beer (4% in the diet) increased hepatic GST activity and reduced carcinogen-DNA adducts in mammary tissue. So far, the beer components responsible for these preventive effects have not been analyzed. Experimental Plant material: An ethanolic hops extract, as well as a CO2-hops extract, was produced, as described in [23], from Humulus lupulus, var. Taurus (Cannabinaceae) and supplied by Hallertauer Hopfenveredlungsgesellschaft (HHV) mbH, Mainburg, Germany. General experimental conditions: NMR spectra were recorded on Bruker Avance 500 and Bruker Avance DRX 500 spectrometers in CD3OD. Mass spectra were measured on a Finnigan MAT 90 mass spectrometer. Extraction and fractionation: An ethanolic hops extract was treated with supercritical carbon-dioxide to remove hops α- and β- acids. This “α-/β-acid free” fraction is a commercially available hops extract that has recently been introduced into the brewing industry for producing xanthohumol-enriched beers [9]. From this extract, 20 g was separated by size exclusion column chromatography using Sephadex LH-20 (column: Ø 5.5 x 120 cm). A step gradient from methanol/dichlormethane 50:50 (v/v), to 70:30 (v/v) to 90:10 (v/v) was performed to obtain the following fractions X01 (5.80 g), X02 (2.75 g), X03 QR induction by humulones and isohumulones Natural Product Communications Vol. 3 (12) 2008 1975 (2.64 g), X04 (1.41g), X05 (0.39g), X06 (0.67g), X07 (0.95 g), X08 (1.23 g), X09 (0.05 g), X10 (0.64 g), X11 (0.38 g), X12 (0.47 g), X13 (0.18 g), X14 (0.21 g), X15 (0.16 g), X16 (0.06 g), X17 (0.03 g), and X18 (0.01 g). Synthesis of humulinones: n-Humulinone and cohumulinone were synthesized by partial synthesis, as described in [10b], starting from pure cohumulone and n-humulone, respectively. Structure elucidation was performed as described above. Spectra were in agreement with published literature [10a,b,e]. An intense bitter taste indicated the presence of bitter acids in fractions X07 and X08. Because of higher yield, fraction X08 (330 mg) was further separated by semi-preparative HPLC, which was performed on a RP-18ec column (VP 250/4 Nucleosil 100-5 C18Hop, Macherey–Nagel, Düren, Germany) using acetonitrile/water 56:44 (v/v) with 0.05% TFA. The solvent delivery system was a Waters M-45 (Waters, Milford USA). Peaks were detected with a RIDetector (RI-Detector 8110, Bischoff, Leonberg, Germany) and, after 7 min, collected to yield 5.1 mg of X08.A, 7.1 mg of X08.B, 5.0 mg of X08.C, 4.6 mg of X08.D and 3.9 mg of X08.E. Structures were determined by NMR spectroscopy (1H, 13C, HSQC, HMBC) and ESI mass spectrometry in comparison with literature data [10b-d,f]. Isolation of humulones as reference compounds: Cohumulone, n-humulone and adhumulone were isolated from a hops CO2-extract by a modified counter-current separation, as described previously [11]. Identity was confirmed by NMR and mass spectrometry in comparison with literature data [10b, 24a,b]. Determination of potential cancer chemopreventive activities: Experimental details of most test systems utilized in this study are summarized in [7,15]. Briefly, radical scavenging potential was determined photometrically by reaction with 1,1-diphenyl2-picrylhydrazyl (DPPH) free radicals in a microplate format [7]. Induction of NAD(P)H:quinone reductase (EC 1.6.99.2) activity in cultured Hepa1c1c7 cells was assayed as described in [25], monitoring the NADPH-dependent menadiol-mediated reduction of MTT [3-(4,5-dimethylthiazo-2-yl)-2,5-diphenyltetrazolium bromide] to a blue formazan. Inhibition of lipopolysaccharide-induced inducible nitric oxide synthase (iNOS) activity (EC 1.14.13.39) in murine Raw 264.7 macrophages was quantified via the Griess reaction, as described previously [15,19b]. Acknowledgments - We thank Hallertauer Hopfenveredelungs Gesellschaft mbH (HHV), Mainburg for providing the extracts and financial support for this work. We thank Dr M. Biendl for fruitful discussion and Dr S. Boettcher for LCMSmeasurements. References [1] Verzele M. (1986) 100-Years of hop chemistry and its relevance to brewing. Journal of the Institute of Brewing, 92, 32-48. [2] Teuber M, Schmalreck AF. (1973) Membrane leakage in Bacillus subtilis 168 induced by the hop constituents lupulone, humulone, isohumulone and humulinic acid. Archives of Microbiology, 94, 159-171. [3] (a) De Keukeleire D, De Cooman L, Rong H, Heyerick A, Kalita J, Milligan SR. (1999) Functional properties of hop polyphenols. Basic Life Sciences, 66, 739-760; (b) Hughes P. (2000) The significance of iso-α-acids for beer quality - Cambridge prize paper. Journal of the Institute of Brewing, 106, 271-276; (c) De Keukeleire D. (2000) Fundamentals of beer and hop chemistry. Quimica Nova, 23, 108-112. [4] (a) Stevens JF, Page JE. (2004) Xanthohumol and related prenylflavonoids from hops and beer: to your good health! Phytochemistry, 65, 1317-1330; (b) Kondo K. (2004) Beer and health: preventive effects of beer components on lifestyle-related diseases. Biofactors, 22, 303-310; (c) Gerhauser C. (2005) Beer constituents as potential cancer chemopreventive agents. European Journal of Cancer, 41, 1941-1954. [5] Lee JC, Kundu JK, Hwang DM, Na HK, Surh YJ. (2007) Humulone inhibits phorbol ester-induced COX-2 expression in mouse skin by blocking activation of NF-κB and AP-1: I-κB kinase and c-Jun-N-terminal kinase as respective potential upstream targets. Carcinogenesis, 28, 1491-1498. [6] (a) Nozawa H, Nakao W, Zhao F, Kondo K. (2005) Dietary supplement of isohumulones inhibits the formation of aberrant crypt foci with a concomitant decrease in prostaglandin E2 level in rat colon. Molecular Nutrition and Food Research, 49, 772-778; (b) Shindo S, Tomatsu M, Nakda T, Shibamoto N, Tachibana T, Mori K. (2002) Inhibition of aldose reductase activity by extracts from hops. Journal of the Institute of Brewing, 108, 344-347; (c) Yajima H, Ikeshima E, Shiraki M, Kanaya T, Fujiwara D, Odai H, Tsuboyama-Kasaoka N, Ezaki O, Oikawa S, Kondo K. (2004) Isohumulones, bitter acids derived from hops, activate both peroxisome proliferator-activated receptor α and γ and reduce insulin resistance. Journal of Biological Chemistry, 279, 3345633462; (d) Yajima H, Noguchi T, Ikeshima E, Shiraki M, Kanaya T, Tsuboyama-Kasaoka N, Ezaki O, Oikawa S, Kondo K. (2005) Prevention of diet-induced obesity by dietary isomerized hop extract containing isohumulones, in rodents. International Journal of Obesity (London), 29, 991-997; (e) Miura Y, Hosono M, Oyamada C, Odai H, Oikawa S, Kondo K. (2005) Dietary isohumulones, the bitter components of beer, raise plasma HDL-cholesterol levels and reduce liver cholesterol and triacylglycerol contents similar 1976 Natural Product Communications Vol. 3 (12) 2008 Bohr et al. to PPARalpha activations in C57BL/6 mice. British Journal of Nutrition, 93, 559-567; (f) Shimura M, Hasumi A, Minato T, Hosono M, Miura Y, Mizutani S, Kondo K, Oikawa S, Yoshida A. (2005) Isohumulones modulate blood lipid status through the activation of PPAR alpha. Biochimica et Biophysica Acta, 1736, 51-60; (g) Namikoshi T, Tomita N, Fujimoto S, Haruna Y, Ohzeki M, Komai N, Sasaki T, Yoshida A, Kashihara N. (2007) Isohumulones derived from hops ameliorate renal injury via an antioxidative effect in Dahl salt-sensitive rats. Hypertension Research, 30, 175-184. [7] Gerhauser C, Alt A, Heiss E, Gamal-Eldeen A, Klimo K, Knauft J, Neumann I, Scherf HR, Frank N, Bartsch H, Becker H. (2002) Cancer chemopreventive activity of xanthohumol, a natural product derived from hop. Molecular Cancer Therapeutics, 1, 959-969. [8] Bohr G, Gerhauser C, Knauft J, Zapp J, Becker H. (2005) Anti-inflammatory acylphloroglucinol derivatives from hops (Humulus lupulus). Journal of Natural Products, 68, 1545-1548. [9] Biendl M, Methner F-J, Stettner G, Walker CJ. (2004) Brauversuche mit einem xanthohumolreichen Hopfenprodukt. Brauwelt, 144, 236-244. [10] (a) Meheus J, Verzele M, Alderweireldt F. (1964) Humulinone. Bulletin Des Societes Chimiques Belges, 73, 268-273; (b) Verzele M, De Keukeleire D. (1991) Chemistry and analysis of hop and beer bitter acids. Elsevier Science Publishers B.V., Amsterdem, New York, 1-417; (c) Nord LI, Sorensen SB, Duus JO. (2003) Characterization of reduced iso-α-acids derived from hops (Humulus lupulus) by NMR. Magnetic Resonance in Chemistry, 41, 660-670; (d) Vanhoenacker G, De Keukeleire D, Sandra P. (2004) Analysis of iso-α-acids and reduced iso-α-acids in beer by direct injection and liquid chromatography with ultraviolet absorbance detection or with mass spectrometry. Journal of Chromatography A, 1035, 53-61; (e) Chadwick LR, Nikolic D, Burdette JE, Overk CR, Bolton JL, van Breemen RB, Frohlich R, Fong HH, Farnsworth NR, Pauli GF. (2004) Estrogens and congeners from spent hops (Humulus lupulus). Journal of Natural Products, 67, 2024-2032; (f) Khatib A, Wilson EG, Kim HK, Supardi M, Choi YH, Verpoorte R. (2007) NMR assignment of iso-α-acids from isomerised extracts of Humulus lupulus L. cones. Phytochemical Analysis, 18, 371-377. [11] Hermans-Lokkerbol ACJ, Verpoorte R. (1994) Preparative separation and isolation of three α-bitter acids from hop, Humulus lupulus L, by centrifugal partition chromatography. Journal of Chromatography A, 664, 45-53. [12] Sporn MB, Newton DL. (1979) Chemoprevention of cancer with retinoids. Federation Proceedings, 38, 2528-2534. [13] Valko M, Leibfritz D, Moncol J, Cronin MT, Mazur M, Telser J. (2007) Free radicals and antioxidants in normal physiological functions and human disease. International Journal of Biochemistry and Cell Biology, 39, 44-84. [14] Tagashira M, Watanabe M, Uemitsu N. (1995) Antioxidative activity of hop bitter acids and their analogues. Bioscience Biotechnology and Biochemistry, 59, 740-742. [15] Gerhauser C, Klimo K, Heiss E, Neumann I, Gamal-Eldeen A, Knauft J, Liu GY, Sitthimonchai S, Frank N. (2003) Mechanismbased in vitro screening of potential cancer chemopreventive agents. Mutation Research, 523-524, 163-172. [16] Jaiswal AK. (2000) Regulation of genes encoding NAD(P)H:quinone oxidoreductases. Free Radical Biology and Medicine, 29, 254-262. [17] Eggler AL, Gay KA, Mesecar AD. (2008) Molecular mechanisms of natural products in chemoprevention: induction of cytoprotective enzymes by Nrf2. Molecular Nutrition and Food Research, 52 (Suppl 1): S84-94. [18] Ohshima H, Bartsch H. (1994) Chronic infections and inflammatory processes as cancer risk-factors - Possible role of nitric-oxide in carcinogenesis. Mutation Research, 305, 253-264. [19] (a) Gerhauser C, Heiss E, Herhaus C, Klimo K. (2001) Potential chemopreventive mechanisms of chalcones. In: Dietary anticarcinogens and antimutagens: chemical and biological aspects Vol. 4.4, Johnson IT, Fenwick GR (Eds.), Royal Society of Chemistry, Cambridge, 189-192; (b) Heiss E, Herhaus C, Klimo K, Bartsch H, Gerhauser C. (2001) Nuclear factor kappa B is a molecular target for sulforaphane-mediated anti-inflammatory mechanisms. Journal of Biological Chemistry, 276, 32008-32015; (c) Dinkova-Kostova AT, Liby KT, Stephenson KK, Holtzclaw WD, Gao X, Suh N, Williams C, Risingsong R, Honda T, Gribble GW, Sporn MB, Talalay P. (2005) Extremely potent triterpenoid inducers of the phase 2 response: correlations of protection against oxidant and inflammatory stress. Proceedings of the National Academy of Sciences of the United States of America, 102, 4584-4589. [20] Yamamoto K, Wang J, Yamamoto S, Tobe H. (2000) Suppression of cyclooxygenase-2 gene transcription by humulone of beer hop extract studied with reference to glucocorticoid. FEBS Letters, 465, 103-106. [21] Zhao F, Nozawa H, Daikonnya A, Kondo K, Kitanaka S. (2003) Inhibitors of nitric oxide production from hops (Humulus lupulus L.). Biological & Pharmaceutical Bulletin, 26, 61-65. [22] Nozawa H, Nakao W, Takata J, Arimoto-Kobayashi S, Kondo K. (2006) Inhibition of PhIP-induced mammary carcinogenesis in female rats by ingestion of freeze-dried beer. Cancer Letters, 235, 121-129. [23] Carl H. (1997) Hops and Hop Products (Manual of good practice). EBC Technology and Engineering Forum, Nürnberg, pp. 67-68. [24] (a) Pusecker K, Albert K, Bayer E. (1999) Investigation of hop and beer bitter acids by coupling of high-performance liquid chromatography to nuclear magnetic resonance spectroscopy. Journal of Chromatography A, 836, 245-252; (b) Hoek AC, Hermans-Lokkerbol ACJ, Verpoorte R. (2001) An improved NMR method for the quantification of alpha-acids in hops and hop products. Phytochemical Analysis, 12, 53-57. [25] Prochaska HJ, Santamaria AB. (1988) Direct measurement of NAD(P)H:quinone reductase from cells cultured in microtiter wells: a screening assay for anticarcinogenic enzyme inducers. Analytical Biochemistry, 169, 328-336. NPC 2008 Vol. 3 No. 12 1977 - 1980 Natural Product Communications A Polar Cannabinoid from Cannabis sativa var. Carma Giovanni Appendinoa*, Anna Gianaa, Simon Gibbonsb, Massimo Maffeic, Giorgio Gnavic, Gianpaolo Grassid and Olov Sternere* a Dipartimento di Scienze Chimiche, Alimentari, Farmaceutiche e Farmacologiche, Via Bovio 6, 28100 Novara, Italy b Centre for Pharmacognosy and Phytotherapy, The School of Pharmacy, University of London, 29-39 Brunswick Square, London WC1N 1AX, UK c Dipartimento di Biologia Vegetale e Centro di Eccellenza CEBIOVEM, Università di Torino, Viale Mattioli 25, 10125 Torino, Italy d CRA-CIN Centro di Ricerca per le Colture Industriali, Sede Distaccata di Rovigo, Via Amendola 82, 45100 Rovigo, Italy e Department of Organic Chemistry, Lund University, P.O. Box 124, 221 00 Lund, Sweden appendino@pharm.unipmn.it; Olov.Sterner@organic.lu.se Received: July 29th, 2008; Accepted: October 15th, 2008 The aerial parts of Cannabis sativa var. Carma afforded a novel polar cannabinoid whose structure was established as rac-6’,7’-dihydro,6’,7’-dihydroxycannabigerol (carmagerol, 1) on the basis of spectroscopic data and semisynthesis from cannabigerol (2a). The dihydroxylation of the ω-double bond was detrimental to the anti-bacterial activity. Keywords: Cannabis sativa, Cannabinaceae, carmagerol, cannabigerol, antibacterial activity. The successful development of Sativex, a combination of Cannabis extracts, for the management of multiple sclerosis and cancer pain [1] has rekindled interest in the phytochemistry of Cannabis sativa L. Over 70 natural cannabinoids are known [2], most of them characterized in the 1960s and 70s in the wake of the identification of Δ9tetrahydrocannabinol (THC) as the psychotropic constituent of marijuana [3]. Recently, a series of very apolar terpenyl esters of pre-cannabinoids was reported from a THC-rich chemotype of marijuana [4], suggesting that the earlier investigations, focused on a defined range of polarity, might have missed minor compounds with higher or lower polarity than the major cannabinoids. We report here the isolation of the novel polar cannabinoid 1 from the Carma variety of hemp. This hemp is named after the Piedmontese town of Carmagnola, where the cultivation of the celebrated pest- and stress resistant homonymous fibre hemp thrived for centuries [5]. 9' HO 8' 10' 7' 3' 5' 6' OH 4' OH 1' 2' 2 1 HO 6 4 1 4'' 2'' 5 3 1'' 3'' 5'' OR RO R 2a H 2b Ac The minor (0.0045% isolation yield) cannabinoid 1 was named carmagerol because of its plant origin and its structrual relationship with cannabigerol (CBG, 2a). It was obtained from the polar fraction of an acetone extract from the aerial parts of Carma hemp. Its isolation involved filtration over RP-18 silica gel to eliminate fats and pigments, partition between aqueous methanol and light petroleum to remove most of CBG (2a), its major cannabinoid, and gravity 1978 Natural Product Communications Vol. 3 (12) 2008 column chromatography on silica gel. The 1H NMR spectrum of a fraction more polar than those containing the prenylated flavonoids cannflavins [6] showed a series of signals in the aliphatic region that suggested the presence of a compound with a gross cannabinoid structure. A pure sample was eventually obtained after preparative HPLC on silica gel. Carmagerol (1) was obtained as an optically inactive, colorless gum, and had molecular formula C21H34O4 (MS). Its 1H NMR spectrum was similar to that of CBG (2a) [7], except for the replacement of the terminal olefinic double bond with two oxygenated functions, as evidenced by the observation of an oxymethine resonance at δ 3.35, and by the upfield shift of the allylic geminal methyls, resonating as sharp singlets at δ 1.19 and 1.15. While the chemical shift of the oxymethine was compatible with the epoxidation of the ω-double bond of the geranyl moiety of CBG, the 13C chemical shift of the corresponding carbon (δ 78.2) and of that of the adjacent quaternary oxygenated carbon (δ 73.2) showed that, in accordance with the molecular formula, the terminal double bond of the geranyl residue had undergone dihydroxylation and not epoxidation. To confirm the structure elucidation of carmagerol and obtain further amounts of the compound necessary to investigate its biological profile, a semisynthesis from CBG was undertaken. Racemic dihydroxylation with the Upjohn protocol gave a complex mixture, but, after acetylation, the asymmetric version of the reaction with AD-mix-α [8] afforded, in excellent yield, a compound identical, apart from the optical rotation, to the natural product. A compound with the gross formula of carmagerol was mentioned in a study on the mammal metabolism of cannabigerol. The major metabolic pathway was the hydroxylation of the allylic methyls, but epoxidation of the ω-double bond was also observed, without, however, detecting its hydrolysis product, namely carmagerol [9]. The racemic nature of carmagerol is puzzling, since biological oxidations are generally enantioselective. On the other hand, auto-oxidation of geranylated phenols shows a strong bias toward the proximal, and not the terminal, double bond [10], and carmagerol was clearly detectable by HPLC in crude extracts of Carma hemps (see Experimental), in accordance with its natural origin. Appendino et al. While cannabigerol (2a) is a potent antibacterial agent, especially against the so called super-bugs (IC50 = 1 μg/mL against methicillin-resistant Staphylococcus aureus SA1199B) [11], the activity of carmagerol (1) was modest (IC50 = 32 μg/mL), showing that dihydroxylation of the ω-double bond is detrimental to antibiotic activity, an important observation that points to the existence of strict structure-activity relationships within the antimicrobial cannabinoid chemotype. The characterization of a novel polar cannabinoid from Cannabis sativa suggests that, despite studies spanning almost 50 years and the identification of over 500 different constituents [2], modern phytochemical techniques can still lead to the isolation of new minor compounds missed by earlier studies and worth investigation in terms of bioactivity. Experimental Plant material: Cannabis sativa var. Carma came from greenhouse cultivation at CRA-CIN, Rovigo (Italy), where a voucher specimen is kept, and was collected in November 2006. The isolation and manipulation of all cannabinoids was in accordance with their legal status (Licence SP/101 of the Ministero della Salute, Rome, Italy). Isolation of 1: The powdered plant material (500 g) was distributed as a thin layer on cardboard, and heated at 120°C for 2 h in a ventilated oven to affect decarboxylation, and then extracted with acetone (ratio solvent: plant material 3:1, x 3). The residue (20.5 g) was dissolved in methanol, adsorbed onto a pad of RP-18 silica gel (100 g), and washed with methanol (400 mL). The yellowish filtrate was evaporated and then partitioned between 10% aq. methanol (150 mL) and light petroleum (150 mL). The lower phase was separated, washed again with light petroleum (100 mL), and evaporated The residue (5.1 g) was purified by gravity column chromatograpy on silica gel (50 g), using a light petroleum-EtOAc gradient. Fractions eluted with light petroleum-EtOAc 4:6 were further purified by prep HPLC on a silica gel column (250 x 21.2 mm Chromasyl column) using light petroleum-EtOAc 3:7 as eluant to give 21 mg (0.0042%) 1a as a colorless foam. Cannabinoids from Cannabis sativa Natural Product Communications Vol. 3 (12) 2008 1979 Carmagerol nabigerol, 1) petroleum-EtOAc 7:3, and filtered over silica gel (10 mL). The filtrate was evaporated, dissolved in THF (10 mL), and treated with pyrrolidine (3 mL, 24 mmol, 12 mol. equiv.). After heating at 50°C for 16 h, the reaction was cooled to room temp. and worked up by partition between 2N H2SO4 and EtOAc. The organic phase was separated, washed with brine, dried (Na2SO4), evaporated, and the residue purified by gravity column chromatography on silica gel (10 g, light petroleum-EtOAc 5:5 as eluant) to afford 700 mg 1a (overall 63% from 2a) as a colorless foam. [α]D = +51 (c 0.8, MeOH). (6’,7’-Dihydro-6’-7’-dihydroxycan- Colorless gum. Rf : 0.35 (light petroleum-EtOAc 4:6). IR (KBr): 3293, 3199, 1703, 1601, 1425, 1372, 1155, 1029, 833 cm-1. 1 H NMR (500 MHz, CDCl3): 0.89 (3H, t, J = 7.0 Hz, H-5’’), 1.15 (3H, s, H-10’), 1.19 (3H, s, H-8’), 1.29 (2H, m, H-3’’a,b), 1.31 (2H, m, H-4’’a,b), 1.44 (1H, m, H-5’a), 1.56 (2H, m, H-2’’a,b), 1.60 (1H, m, H-5’b), 1.82 (3H, s, H-9’), 2.13 (1H, m, H-4’a), 2.27 (1H, m, H-4’b), 2.44 (2H, t, J = 7.8 Hz, H-1’’a,b), 3.35 (1H, dd, J = 10.2, 2.1 Hz, H-6’), 3.38 (2H, t, J = 7.2 Hz, H-1’), 5.32 (1H, br t, J = 7.2 Hz, H-2’), 6.24 (2H, s, H-4 and H-6). 13 C NMR (125 MHz, CDCl3): 14.0 (t, C-5’’), 16.1 (q, C-9’), 22.2 (t, C-1’), 22.5 (t, C-4’’), 23.3 (q, C-10’), 26.3 (q, C-8’), 29.4 (t, C-5’), 30.8 (t, C-2’’), 31.4 (t, C-3’’), 35.5 (t, C-1’’), 36.9 (t, C-4’), 73.2 (s, C-7’), 78.2 (d, C-6’), 108.2 (d, C-4 and C-6), 110.9 (s, C-2), 122.8 (d, C-2’), 139.4 (s, C-3’), 146.2 (s, C-5), 154.8 (s, C-1 and C-3). CI-EIMS: m/z [M]+ 373 (M + Na)+, (C21H34O4 + 23)+. Synthesis of carmagerol (1) from cannabigerol (2a): To a stirred soln of AD-mix-α (3.5 g) in tert-ButOHwater (1:1, 10 mL), N-methylmorpholine oxide (875 mg, 7.5 mmol, 3 mol. equiv.), methansulfonamide (250 mg, 2.5 mmol, 1 mol. equiv.), and a soln of cannabigerol diacetate (2b, 1.0 g, 2.5 mmol; obtained from the treatment of 2a (1 g) with excess Ac2O (10 mL) in pyridine (10 mL) in tert-ButOH (7 mL) were added. After stirring at room temp. overnight, the reaction was worked up by the addition of sat. Na2SO3, and stirred at room temp. for 30 min. EtOAc was then added, and the reaction mixture was extracted with EtOAc. The organic phase was washed with brine, dried with Na2SO4, and evaporated. The residue was taken up in light Analysis of carmagerol in Cannabis sativa var. Carma: A 5 g sample of plant material was extracted with acetone (3 x 30 mL) by stirring at room temp. (10 min. for each extraction). The pooled extracts were evaporated, and a portion of the residue (100 mg) was dissolved in 1 mL methanol and filtered in a Pasteur pipette over Celite (50 mg). The filtration pad was washed with 0.5 mL methanol, and the pooled filtrates were diluted with 0.15 mL water. After washing twice with light petroleum (2 x 2.5 mL) to remove most of the apolar cannabinoids, the lower methanol phase was evaporated, taken up in 200 mL methanol, and analyzed by HPLC (C-18 column, detection at 210 nm). The following conditions were employed: Solvent A: 0.5% orthophosphoric acid in water; Solvent B: acetonitrile. Gradient: from 0 to 8 min, 60% A, 40% B; from 9 to 14 min, 50% A, 50% B, from 15 to 24 min 10% A, 90% B, from 25 to 30 min 1% A and 99% B. The Rt of 1 was 13.8 min, and its concentration was in the range of 56-98 mg/Kg depending on the sample analyzed Acknowledgments – We are grateful to Dr Lucia Maxia (Università del Piemonte Orientale, Faculty of Pharmacy) for her help in the isolation of carmagerol. References [1] Perez J, Ribera MV. (2008) Managing neuropathic pain with Sativex: a review of pros and cons. Expert Opinions in Pharmacotherapy, 9, 1189-1195. [2] El-Sohly MA, Slade D. (2005) Chemical constituents from Marijuana: The complex mixture of natural cannabinoids. Life Sciences, 78, 539-548. [3] Mechoulam R, Gaoni Y (1964) Isolation, structure and partial synthesis of an active constituent of hashish. Journal of American Chemical Society, 86, 1646-1647. [4] Ahmed SA, Ross SA, Slade D, Radwan MM, Zulfiquar F, Elsohly MA. (2008) Cannabinoid ester constituents from high-potency Cannabis sativa. Journal of Natural Products, 71, 536-542. [5] The name of the famous French revolutionary song “la Carmagnole” was in fact inspired by the trade of this variety of hemp, characterized by very long fibers and insuperable for the manufacture of naval ropes, from Carmagnola and Marseille. See: http://architettura.supereva.com/image/festival/1998/en/bonino.htm 1980 Natural Product Communications Vol. 3 (12) 2008 Appendino et al. [6] Minassi A, Giana A, Ech Chahad A, Appendino G. (2008) A regiodivergent synthesis of ring C prenylflavones. Organic Letters, 10, 2257-2270. [7] Choi YH, Hazekamp A, Peltenburg-Looman AMG, Frédérich M, Erkelens C, Lefeber AWM, Verpoorte R. (2004) NMR assignments of the major cannabinoids and cannabiflavonoids isolated from flowers of Cannabis sativa. Phytochemical Analysis, 15, 345-354. [8] For a related example, see: Vidari G, Di Rosa A, Castronovo F, Zanoni G. (2000) Enantioselective synthesis of each stereoisomer of the pyranoid linalool oxides: the geraniol route. Tetrahedron Asymmetry, 11, 981-989. [9] Harvey, D, Brown NK. (1990) In vitro metabolism of cannabigerol in several mammalian species. Biomedical & Environmental Mass Spectrometry, 19, 545-553. [10] A series of cannabigerol derivatives epoxidized on the proximal double bond have recently been reported (Radwan MM, Ross SA, Slade D, Ahmed SA, Zulfiquar F, Elsohly MA. (2008) Isolation and characterization of new Cannabis constituents from a high potency variety, Planta Medica, 74, 267-272. [11] Appendino G, Gibbons S, Giana A, Pagani A, Grassi G, Stavri M, Smith E, Mukhlesur Rahman M. (2008) Antibacterial cannabinoids from Cannabis sativa. A structure-activity study. Journal of Natural Products, 71, 1427-1430. NPC 2008 Vol. 3 No. 12 1981 - 1984 Natural Product Communications HPLC-DAD-MS Fingerprint of Andrographis paniculata (Burn. f.) Nees (Acanthaceae) Sabrina Arpinia, Nicola Fuzzatia*, Andrea Gioria, Emanuela Martinob, Giacomo Mombellia, Luca Pagni a and Giuseppe Ramaschi a. a Indena S.p.A. Research and Development Laboratories, Via Don Minzoni 6, 20090 Settala (MI), Italy b Dipartimento di Ecologia del Territorio, Università di Pavia, Via S. Epifanio 14, 27100 Pavia, Italy nicola.fuzzati@indena.com Received: July 1st, 2008; Accepted: October 24th, 2008 An HPLC-UV fingerprint analysis was developed for the quality evaluation of Andrographis paniculata aerial parts. HPLC-DAD-MS experiments allowed the identification of eleven diterpenes and five flavonoids. Plant material of Indian and Chinese origin was evaluated employing the developed method. The chemical fingerprints of the plant material of different origins do not show significant differences. 15 13 13 13 13 13 11 12 12 12 12 7 8 9+ 10 6 5 3 2 12 16 14 A 0.20 1 AU 0.40 15 11 7 8 6 5 3 2 16 9+10 B 0.20 1 AU 0.40 14 4 0.00 11 7 8 9+ 10 5 3 2 16 C 0.20 1 AU 0.40 14 15 4 0.00 5 6 3 2 15 11 7 8 9+10 16 D 0.20 1 AU 0.40 14 4 0.00 4 0.00 11 7 8 6 2 3 1 5 9+ 10 E 0.20 14 15 16 0.40 AU The aerial parts of Andrographis paniculata (Acanthaceae) have been used widely in Indian folk medicine and Ayurveda for the treatment of different diseases such as dysentery, cholera, diabetes, consumption, influenza, bronchitis and gonorrhoea [1]. Chinese medicine employs this herb mainly for its bitter properties as a treatment for digestive problems and for a variety of febrile illnesses [2]. More recently, A. paniculata standardized extracts have become very popular in Europe for the treatment of upper respiratory infection and influenza [3]. Among these extracts, the product developed by the Swedish Herbal Institute and called Kan Jang is the most widely tested. Kan Jang is standardized to contain 5.25 mg of a mixture of andrographolide and deoxyandrographolide per tablet. Andrographolide is a diterpene lactone which is considered to be the active principle. Hence all the published analytical methods are focused on the determination of andrographolide [4-10]. However, phytochemical investigations of A. paniculata have shown the presence of more than twenty labdane diterpenoids, and over ten 2’-oxygenated flavonoids have been reported. In the present paper a new HPLC method, which allows the detection of both classes of compound in plant material of different origins, is described. Since the greater part of the 4 Keywords: Andrographis paniculata, fingerprint, HPLC-DAD-MS. 0.00 8.00 10.00 12.00 14.00 16.00 18.00 20.00 22.00 24.00 26.00 28.00 Minutes Figure 1: HPLC-UV chromatograms of A. paniculata aerial parts A: batch # 117302 China; B: batch # 114817 India; C: batch # 111364 India; D: batch # 112927 China; E: batch # 112928 China. commercialized A. paniculata aerial parts comes from China and India, plant material of Chinese and Indian origin was analyzed. The HPLC-UV chromatograms, recorded at 220 nm, showed several peaks (Figure 1), but no significant differences between the two origins. 1982 Natural Product Communications Vol. 3 (12) 2008 Fuzzati et al. Table 1: HPLC-DAD-MS data of peaks 1-16. Peak 1 2 Rrt 0.65 0.79 UV max nm Molecular ions m/z Fragments m/z 227 1047 [2M+Na+]. 551 [M+K+] 535[M+Na+] 351 [M +H-Glc]+, 333 [M+H-Glc-H2O]+, 315 [M+H-Glc-2H2O]+, 297, 285, 257 200 739 [2M+K+] 701 [2M+H+]. 389 [M+K+] 351[M+H+] + 3 4 5 0.94 1.00 1.03 200 225 237 sh 286 325 735 [2M+K ] 697 [2M+H+]. 387 [M+K+] 349[M+H+] 739 [2M+K+] 701 [2M+H+] 389 [M+K+] 351[M+H+] + 963 [2M+K ] 501 [M+K+] O O HO O O O O OH 333 [M+H -H2O]+, 315 [M+H -2H2O]+, 297, 285, 257 HO HO OH OR 1 R= glucose 4 R= H andrographiside andrographolide OGlucose 2, 6 14-deoxy-11(or 12)-hydroxyandrographolide 16 neoandrographolide O O O O O OH O O + 331 [M+H -H2O] , 313 [M+H -2H2O]+, 301 HO HO HO 333 [M+H -H2O]+, 315 [M+H -2H2O]+, 297, 285, 257 OR OH OH 7 andropanolide 3 14-deoxy-11-oxoandrographolide 8 R= glucose andropanoside 15 R= H 14-deoxy-andrographolide R3 O O R2 O O O O O O + 301 [M +H-Glc] , OGlucose O 5 andrographidine A OR1 O HO + 6 7 8 9 1.09 200 739 [2M+K ] 389 [M+K+] + 333 [M+H -H2O] , 315 [M+H -2H2O]+, 297 226 739 [2M+K+] 701 [2M+H+]. 389 [M+K+] 351[M+H+] 1.19 200 1031 [2M+K+] 993 [2M+H+]. 535 [M+K+] 497[M+H+] 1.22 991[2M+K+] 975 [2M+Na+]. 515 [M+K+] 499 [M+Na+] 477[M+H+] 315 [M +H-Glc]+, 260, 341 333 [M +H-Glc]+, 315 [M+H-Glc-H2O]+, 297 [M+H-Glc-2H2O]+, 285, 257 1.16 333 [M+H -H2O]+, 315 [M+H -2H2O]+, 297, 285, 257 335 [M +H-Glc]+, 317 [M+H-Glc-H2O]+, 299 [M+H-Glc-2H2O]+, 287, 259 10 1.23 255 1027 [2M+K+] 989 [2M+H+]. 533 [M+K+] 495[M+H+] 11 1.63 266, 344 959 [2M+K+] 499 [M+K+] 299 [M +H-Glc]+ 12 1.43 263, 335 529 [M+K+] 513 [M+Na+] 329 [M +H-Glc]+ 13 1.49 263, 355 1079 [2M+K+] 559 [M+K+] 543 [M+Na+] 359 [M +H-Glc]+, 252 703 [2M+K+] 687 [2M+Na+] 665 [2M+H+]. 371 [M+K+] 333[M+H+] 315 [M+H-H2O]+, 297 [M+H -2H2O]+, 285, 257 204 707 [2M+K+] 691 [2M+Na+] 669 [2M+H+]. 373[M+K+] 357[M+Na+] 317 [M+H-H2O]+, 299 [M+H -2H2O]+, 287, 257 999 [2M+K+] 983 [2M+Na+]. 519 [M+K+] 503[M+Na+] 319 [M +H-Glc]+, 301 [M+H-Glc-H2O]+, 289 14 15 16 1.59 1.61 1.66 201 OR 10 R= glucose 14-deoxy-11,12-didehydroandrographiside 14 R= H 14-deoxy-11,12-didehydroandrographolide 9 11 12 13 R1= H R2= glucose R1= glucose R2= H R1= glucose R2= OCH3 R1= glucose R2= OCH3 R3= H skullcapflavone I 2'-O-glucoside R3= H andrographidine C R3= H andrographidine E R3= OCH3 andrographidine D Figure 2: Structures of compounds 1-16. The HPLC-DAD-UV analysis of A. paniculata extracts allowed differentiation betweeen diterpenes (peaks 1-4, 6-8, 10 and 14-16) and flavonoids (peaks 5, 9 and 11-13). In addition, the ESI-MS of the detected peaks exhibited clusters signals at [2M+K+], [2M+Na+], [2M+H]+ and adduct ions at [M+K+] and [M+Na+] allowing the determination of the molecular weights (Table 1, Figure 2). Furthermore, several fragments attributable to the loss of glucose (Glc) and to successive losses of water were observed. Peaks 1, 4 and 7 exhibited a maximum at approximately 225 nm, indicating the presence of an α,β-unsaturated-γ-lactone group with an exo-cyclic double bond. Peaks 4 and 7 showed the same ESI-MS spectra indicating that they are isomers. Peak 4 was identified as andrographolide by injection of the reference compound. A number of isomers of andrographolide were previously described [8,11-13], such as 14−epi-andrographolide, isoandrographolide and andropanolide, all possessing the α,βunsaturated-γ-lactone group with an exo-cyclic double bond. In a previous work [8], isoandrographolide exhibited the same HPLC behaviour as peak 7. However, isoandrographolide was described with two different structures: A [8,12] and B [11,13] (Figure 3). Only recently the correct structure and stereochemistry of HPLC-UV fingerprint analysis of Andrographis paniculata O O 16 14 OH O OH O 12 20 11 1 3 HO 17 9 5 6 HO 18 19 OH OH 14-epi-andrographolide isoandrographolide structure A O O O O OH Natural Product Communications Vol. 3 (12) 2008 1983 The ESI-MS of the flavonoids (peaks 5, 9 and 11-13) showed signals due to the loss of a glucose unit allowing the identification of the aglycons. Comparison of the UV and MS data with those of the literature allowed the identification of peak 5 as andrographidine A [15], peak 9 as skullcapflavone I2’-glucoside [16], peak 11 as andrographidine C [15], peak 12 as andrographidine E [15] and peak 13 as andrographidine D [15]. H H O HO HO OH OH andropanolide isoandrographolide structure B Figure 3 isoandrographolide (structure B) was determined by X-ray analysis [13]. In this paper, the authors isolated also a compound which exhibits the same 13C NMR signals as the isoandrographolide structure A described in the work of Li and Fizloff [8]. Furthermore, they determined the correct stereochemistry of the compound and named it andropanolide. Hence peak 7 should be identified as andropanolide. The ESI-MS of peak 1 exhibited the loss of a glucose unit and fragments attributable to the andrographolide moiety. Peak 1 was identified as andrographiside [12]. The ESI-MS of peaks 2 and 6 suggested that these compounds are isomers of andrographolide. However, their UV spectra showed no absorption at 220 nm indicating the presence of an α,β-unsaturated-γ-lactone group with an endo cyclic double bond. Peaks 2 and 6 were identified as the compounds previously described by Matsuda et al. [12] and named 14-deoxy-11(or 12)hydroxyandrographolide. Peaks 3, 8, 15 and 16 exhibited UV spectra similar to those of peaks 2 and 6. The study of their ESI-MS allowed the identification of peak 3 as 14-deoxy-11oxoandrographolide [14], peak 8 as andropanoside, peak 15 as 14-deoxyandrographolide and peak 16 as neoandrographolide [12]. Peaks 10 and 14 exhibited a maximum at approximately 250 nm indicating the presence of two conjugated double bonds. The study of their ESI-MS allowed the identification of peak 10 as 14-deoxy-11,12-didehydroandrographiside [12], and peak 14 as 14-deoxy-11,12-didehydroandrographolide [12]. Experimental Plant material: A. paniculata aerial parts (batches # 111364 and 114817) were collected in November 2005 in Maredehalli (India). Batches # 112927, 112928 and 117302 were purchased from Yee Po International Co. (Hong Kong, China). Voucher specimens are kept at Indena R&D Laboratories, Settala, Italy. Plant material extraction: About 2.5 g of A. paniculata plant material, milled through a 6 mm screen, was weighted into a 250 mL Pyrex flask. After addition of 120 mL ethanol, 80% v/v, the samples were shaken in a mechanical shaker for 3 h at 60°C. The suspension was filtered through paper and the extraction procedure repeated. The obtained extracts were filtered and pooled in a 250 mL volumetric flask and finally diluted to volume with ethanol 80% v/v. Reference material: Andrographolide 98% (catalog number # 365645) was purchased from SigmaAldrich (Milano, Italy). HPLC-DAD-MS analyses: The HPLC-DAD-MS analyses were conducted on a Finnigan Surveyor HPLC system equipped with a SpectraSYSTEM DAD UV6000LP and a Finnigan MAT LCQ ion trap mass spectrometer fitted with a Microsoft® Window® XP™ data system and an ESI interface. The chromatographic separation was performed on a column of Zorbax SB-C18 (5 µm) (250 x 4.6 mm I.D.) from Agilent Technologies, employing water (solvent A) and methanol (solvent B). The linear gradient applied started from 60% A going to 30% A in 30 min, then in 1 min to 10% A, finally remaining 9 min at 10% A. The column temperature was set at 20°C and the flow rate at 1 mL/min. The injected volume was 10 μL. The wave length range of the DAD detector was set at 210-400 nm. Mass spectrometer conditions were optimized in order to 1984 Natural Product Communications Vol. 3 (12) 2008 achieve maximum sensitivity. ESI conditions: source voltage 6.0 kV, sheath gas flow rate 70 au, source current 80 μA, capillary voltage 34 V and capillary temperature 240°C. Full scan spectra from 150 to Fuzzati et al. 1500 u in the positive ion mode were obtained (scan time 1 s). Ion trap conditions: acquisition in automatic gain control with a max-inject time of 200 msec. References [1] (a) Gupta AK, Neeraj Tandan. (2004) Review on Indian Medicinal Plants Vol. 2. Indian Council of Medical Research, New Delhi, 283-306; (b) Perumal Samy R, Thwin MM, Gopalakrishnakone P. (2007) Phytochemistry, pharmacology and clinical use of Andrographis paniculata. Natural Product Communications, 2, 607-618. [2] Pharmacopoeia of the People’s Republic of China 2005 (English Edition). People’s Medical Publishing House, Beijing, 121. [3] Kligler B, Ulbricht C, Basch E, De Franco Kirkwood C, Rae Abrams T, Miranda M, Khalsa KPS, Giles M, Boon H, Woods J. (2006) Andrographis paniculata for the treatment of upper respiratory infection: a systematic review by the natural standard reserch collaboration. Explore, 2, 25-29. [4] Sharma A, Lal K, Handa SS. (1992) Standardization of the Indian crude drug Kalmegh by high pressure liquid chromatographic determination of andrographolide. Phytochemical Analysis, 3, 129-131. [5] Jain DC, Gupta MM, Saxena S, Kumar S. (2000) LC analysis of hepatoprotective diterpenoids from Andrographis paniculata. Journal of Pharmaceutical and Biomedical Analysis, 22, 705-709. [6] Li W, Fitzloff JF. (2002) Determination of andrographolide in commercial andrographis (Andrographis paniculata) products using HPLC with evaporative light scattering detection. Journal of Liquid Chromatogrophy & Related Technologies, 25, 1335-1343. [7] Pholphana N, Rangkadilok N, Thongnest S, Ruchirawat S, Ruchirawat M, Stayavivad J. (2004) Determination and variation of three active diterpenoids in Andrographis paniculata (Burm.f.) Nees. Phytochemical Analysis, 15, 365-371. [8] Li W, Fitzloff JF. (2004) HPLC-PDA determination of bioactive diterpenoids from plant materials and commercial products of Andrographis paniculata. Journal of Liquid Chromatogrophy & Related Technologies, 27, 2407-2420. [9] Srivastava A, Misra H, Verma RK, Gupta MM. (2004) Chemical fingerprinting of Andrographis paniculata using HPLC, HPTLC and densitometry. Phytochemical Analysis, 15, 280-285. [10] Akowuah GA, Zhari I, Norhayati I, Mariam A. (2006) HPLC and HPTLC densitometric determination of andrographolides and antioxidant potential of Andrographis paniculata. Journal of Food Composition and Analysis, 19, 118-126. [11] Cava MP, Chan WR, Stein RP, Willis CR. (1965) Andrographolide. Further transformations and stereochemical evidence; the structure of isoandrographolide. Tetrahedron, 21, 2617-2632. [12] Matsuda T, Kuroyanagi M, Sugiyama S, Umehara K, Ueno A, Nishi K. (1994) Cell differentiation-inducing diterpenes from Andrographis paniculata Nees. Chemical Pharmaceutical Bulletin, 42, 1216-1225. [13] Pramanick S, Banerjee S, Achari B, Das B, Sen Sr. AK, Mukhopadhyay S, Neuman A, Prangé T. (2006) Andropanolide and isoandrographolide, minor diterpenoids from Andrographis paniculata: structure and X-ray crystallographic analysis. Journal of Natural Products, 69, 403-405. [14] Balmain A, Connoly JD, (1973) Minor diterpenoids constituents of Andrographis paniculata Nees. Journal of Chemical Society Perkin Transactions I, 1247-1251 [15] Kuroyanagi M, Sato M, Ueno A, Nishi K. (1987) Flavonoids from Andrographis paniculata. Chemical and Pharmaceutical Bulletin, 11, 4429-4435. [16] Gupta KK, Taneja SC, Dhar KL. (1996) Flavonoid glycoside of Andrographis paniculata. Indian Journal of Chemistry, 35B, 512-513. NPC Natural Product Communications Diterpenoid Alkaloids and Phenol Glycosides from Aconitum naviculare (Brühl) Stapf. 2008 Vol. 3 No. 12 1985 - 1989 Stefano Dall’Acquaa*, Bharat B. Shresthab , Mohan Bikram Gewalic, Pramod Kumar Jhab , Maria Carrarad and Gabbriella Innocenti a a Department of Pharmaceutical Sciences, University of Padova, Padova, Italy b Central Department of Botany, Tribhuvan University, Kirtipur, Kathmandu, Nepal c Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal d Department of Pharmacology and Anesthesiology, University of Padova, Padova, Italy stefano.dallacqua@unipd.it Received: June 20th, 2008; Accepted: October 28th, 2008 Phytochemical investigation of the aerial parts of Aconitum naviculare, a medicinal plant used in traditional Nepalese medicine, led to the isolation and characterization of two new diterpenoid alkaloids, navirine B (1), and navirine C (2), along with (+) chellespontine (3), kaempferol-7-O-β-D-glucopyranosyl(1→3)α-L-rhamnopyranoside (4), kaempferol-7-O α-Lrhamnopyranoside,3-O-β-D-glucopyranoside (5), p-coumaric-4-O-β-D-glucopyranoside acid (6), and ferulic-4-O-β-Dglucopyranoside acid (7). The structures of the isolated compounds were elucidated on the basis of extensive analyses of 1D and 2D NMR spectra (HMQC, HMBC, COSY, ROESY) and HR-MS data. The antiproliferative activity of alkaloids 1-3 against human tumor cell lines (LoVo and 2008) was also evaluated. Keywords: Aconitum naviculare, diterpenoid alkaloids, antiproliferative activity, traditional medicine. Aconitum species are well-known for their contents of C19 and C20 diterpenoid alkaloids. Some of these (e.g., aconitine) are highly toxic and some exhibit powerful biological activities, for example antiarrhythmic [1,2], analgesic [2], antiinflammatory [2], antiepileptic [2] and antiproliferative [3], making them potential new pharmaceutical entities. Aconitum naviculare (Brühl) Stapf (Ranunculaceae) is a biennial medicinal herb of Alpine grassland (>4000 m a.s.l.) found in the trans-Himalayan region of Nepal, i.e., the Manang, Mustang and Dolpa districts. The aerial parts are used in Nepalese and Tibetan folk medicine against cold, fever and headache, as well as for sedative and analgesic remedies [4,5]. In the Manang region, the aerial parts are collected during flowering. The local inhabitants usually dry the whole plant and prepare a bitter decoction, which is used for various medicinal purposes. Although Manangis living in and outside Manang commonly use A. naviculare, it has not yet become an item of trade. Due to its supposed high effectiveness in traditional healthcare, A. naviculare may become a potential source of income for mountain people. Little information is available regarding the phytochemical composition of A. naviculare. Gao et al. [4] reported the isolation of a new diterpenoid alkaloid and some atisine-like alkaloids. We recently reported three novel glycosylated flavonoids from this plant [6]. Here we report the isolation and characterization of two new diterpenoid alkaloids, navirine B (1) and navirine C (2), and five known compounds (3-7) from A. naviculare. The isolated diterpenoid alkaloids 1-3 were also studied for their ability to affect tumor cell proliferation. In particular, antiproliferative activity against ovarian (2008 cells) and colon (LoVo cells) adenocarcinoma was tested. Compound 1 had a molecular formula of C30H40N2O3 on the basis of the protonated molecular ion [M+H]+ displayed at m/z 477.3122 in the HRAPITOFMS. The IR spectrum showed absorption bands supporting the presence of an imino group (1670 cm-1). 1986 Natural Product Communications Vol. 3 (12) 2008 Dall’Acqua et al. 12 20 2 N 3 19 11 1 14 9 10 5 4 H OH 6 18 13 13 13 12 17 OH 11 O 16 1' 2' 15 2 6' 8 3' 5' 7 4' 1 7' N 3 1 4 19 8' N 15 10 5 H 18 H 8 1' 1 2' H 5' 4' 7' 2 3 O 10 5 N 22 4 19 H 18 16 14 9 2 6' 3' 7 6 11 O 16 14 9 17 12 17 H8 15 OH 7 6 3 8' N 9' 9' 9' 9' Figure 1: Structure of isolated compounds 1-3. The 1H NMR spectrum showed signals ascribable to one methyl group (δ 1.05 s, 3H), an N,N-dimethyl group (δ 2.40 s, 6H), and one olefinic proton (δ 5.67 brs, 1H). Two ortho coupled doublets (J = 8.0) in the aromatic region (δ 6.85 and 7.12, 2H each) also indicated the presence of an o-p disubstituted aromatic ring. The structure of compound 1, a diterpenoid alkaloid, was obtained by exhaustive analysis of COSY, HMQC and HMBC data. Various spin systems were detected when COSY and HMQC data were compared, establishing the connectivity between CH2 in positions 1,2,3 and 6,7 and between positions 9,11,12,13. Further connectivity between the highly deshielded CH-19 (δH 7.43 s, 1H; δC 169.9) and CH-20 (δH 3.57 brs, 1H; δC 80.6) and also between the olefinic CH-15 (δH 5.67 brs, 1H; δC 130.8) and CH2-17 (δH 4.55 brs, 1H; δC 68.8) were seen in the COSY spectrum. Diagnostic long-range (HMBC) correlations were observed from the methyl group 18 and carbon resonances at δ 30.9 (C-3), 44.9 (C-4), 72.6 (C-5) and 169.9 (C-19). HMBC correlations, observed from the proton signal of CH-20 (δ 3.57) with carbon resonances C-19 and C-5, supported the presence of a six-member ring containing an imino group. Diagnostic long-range correlations were observed from the same proton signal (H-20) with C-1 (δ 27.9), C-9 (47.1), C-5 (72.6) and C-8 (43.8). HMBC correlations of the proton at δ 5.67 (H-15) with C-9 (δ 47.1), C-12 (δ 31.7) and C-17 (68.8) were also seen. These observations, as well as analysis of COSY and HMQC data, suggested two more hexa-atomic rings in the compound. Long-range correlations observed from proton signals at δ 1.91-1.56 (CH2-13) with carbon resonances at δ 146.8 (C-16) and 80.6 (C-20) supported the linkage between positions 20 and 14. Long-range correlations observed from the aromatic doublet δH 7.12 (H-3’ 5’) with the carbon at δC 33.3 (C-7’) and from the methyl group at δH 2.59 (CH3-9’) with the carbon at δC 52.5 (C-8’) suggested the presence of one hordenine moiety in the molecule. NOESY data and comparisons with the spectral data of Gao et al. [4] for navirine evidenced the relative stereochemistry of the molecule, supporting an α orientation for groups at positions 10, 8 and 12. NOESY cross-peaks were also obtained between the exchangeable proton signal at δ 3.48 (OH-5) and H-9 (δ 1.64), which were assigned as β on the basis of previous references [4,7], suggesting a β orientation for the hydroxy group. With all this evidence, compound 1 was established as a new alkaloid, called navirine B (5β-hydroxy navirine). The HRAPITOFMS of compound 2 displayed a protonated molecular ion [M+H]+ at m/z 461.3532, corresponding to a molecular formula of C31H45N2O. The 1H NMR spectrum of compound 2 was quite similar to that of compound 1, but there were some differences. It lacked the signal for the imino proton, and the proton signal of H-20 was shifted downfield to δ 2.53 (δ 3.57 for 1). In addition, a singlet signal integrating for nine protons was observed at δ 2.50, supporting the presence of three nitrogen linked methyl groups. Extensive analyses of COSY, HMQC and HMBC data revealed a similar diterpenoid skeleton, as well as the same hordenine moiety as found in compound 1. In the HMBC spectrum, long-range correlations were observed between the methyl group at δ 1.00 (CH3-18) and carbon resonances at δ 34.1 (C-3), 41.7 (C-4), 53.5 (C-5) and 58.0 (C-19), which supported the presence of aminolinked CH2 at position 19 (δH 2.30; δC 58.0), as well as a CH group at position 5 (δH 1.26; δC 53.5). The β configuration for H-5 was established on the basis of the NOESY correlation observed from the methyl group at δ 1.00 and the proton signal at δ 1.26 (H-5). Complete analyses of 1D and 2D NMR data afforded a novel structure, called navirine C. Diterpenoid alkaloids and glycosides from Aconitum naviculare Natural Product Communications Vol. 3 (12) 2008 1987 Compound 3 showed a protonated molecular ion [M+H]+ at m/z 344.2590, establishing its molecular formula as C22H33NO2. The 1H NMR spectrum of 3 displayed one aldehydic proton signal (8.71 s, 1H), one exocyclic methylene (δ 5.02 d, 5.12 d, 1H each; J = 1.0 Hz), one methyne signal linked to an electronegative atom (δ 3.70 m, 1H), and one methyl group (δ 1.06 s, 3H). Its COSY, HMQC and HMBC data revealed a diterpenoid skeleton. Compound 3 possessed a hydroxyl group at position 15, as supported by the HMBC correlation between H-17 (δ 5.05 and 5.12) and the carbon resonance at δ 74.7 (C-15). The β orientation of the OH group at position 15 was deduced on the basis of NOESY cross-peaks observed between H-15 and H-13 and H-14. The proton and carbon resonances at position 15 were at δH 3.70 and δC 74.7 respectively, due to the β-OH group. The H 9 configuration in compound 3 was established by observing the NOESY correlations from the methyl group 18 (δ 1.06) and H-5 (δ 1.38), and from this latter and the H-9 signal (δ 2.19). On the basis of the spectral data, compound 3 was established as chellespontine [8]. whereas, in 2008 cells, only compound 1 induced a marked inhibition of cell growth, as shown in Table 1 as IC50 values. To our knowledge alkaloid 3, flavonol glycosides 4 and 5, and phenylpropanoid glycosides 6 and 7 have been isolated and characterized from A. naviculare for the first time. Recently some Aconitum alkaloids have been evaluated for their antiproliferative activity against A172 malignant cells [3]. For this reason, we decided to study this activity for the isolated diterpenoid alkaloids 1-3. Our study on the phytochemical composition of A. naviculare may be useful in improving scientific knowledge of this Nepalese medicinal plant. Experimental Optical rotations were measured on a Jasco P2000 digital polarimeter, IR spectra on a Perkin Elmer 1600 FT-IR spectrometer, and NMR spectra, in CDCl3 or CD3OD, on a Bruker AMX-300 spectrometer, operating at 300.13 MHz for 1H NMR and 75.03 MHz for 13C NMR. 2D experiments, 1H-1H DQF-COSY, and inverse-detected 1H-13C HMQC and HMBC spectra were performed with UXNMR software. HRMS were obtained on an API-TOF spectrometer (Mariner Biosystems). Samples were diluted in a mixture of H2O-AcCN (1:1), with 0.1% formic acid for positive ion mode, and directly injected at a flow rate of 10 µL/min. Sephadex LH 20 and silica gel 60 were used for column chromatography. Silica gel plates were used for preparative and analytical TLC (Merck cat. 5717 and 5715). Compounds on TLC were detected with a UV lamp (254 nm) and by treating plates with Dragendorff’s reagent. Semi-preparative HPLC was performed on a Gilson series 305 liquid chromatograph equipped with a LiChrosphere 100 RP-18 column (particle size 10 μm, 250 x 10 mm ID, E. Merck). Table 1: Antiproliferative activity of compounds 1-3. LoVo cellsa IC50 (confidence limits) μM 22 (19-25) 2008 cellsa IC50 (confidence limits) μM 33 (30-37) 2 nd nd 3 38 (33-41) nd 33.3 (28.7-36.1) 13.8 (11.5-!6.5) Compound 1 Cisplatin b a LoVo: colon cell line; 2008: ovarian cell line. Cisplatin was used as a reference compound. nd: IC50 is not determined. b Compounds 1 and 3 showed a significant antiproliferative activity, whereas compound 2 was inactive. The capacity of the compounds to affect tumor cell growth revealed a dose-dependent effect. In particular, colon cell line LoVo was more sensitive than ovarian cells 2008. In fact, compounds 1 and 3 were able to decrease cell proliferation in LoVo cells, Plant material: Aerial parts, including stems, leaves, flowers and immature fruits of Aconitum naviculare were collected from Ladtar (4100 m a.s.l., upper Manang) during the last week of September 2004. A voucher specimen was collected during a field survey and deposited at Tribhuvan University Central Herbarium (TUCH; n° ANV904); identification was confirmed by Prof. Ram Prasad Chaudhary of the Central Department of Botany, Tribhuvan University, Kathmandu. Extraction and isolation: Air-dried powdered aerial parts (100 g) were exhaustively extracted in a Soxhlet apparatus with MeOH. The solvent was evaporated under reduced pressure (230–250 mbar) and a semisolid MeOH extract was obtained (8 g). About 1988 Natural Product Communications Vol. 3 (12) 2008 5.0 g of this extract was suspended in a mixture of 9:1 H2O–MeOH (200 mL), and the pH was adjusted to 2 with 5% aq HCl. The solution was then partitioned first with CHCl3 (5 x 50 mL) and then with EtOAc (5 x 50 mL). Solvents were removed under vacuum, yielding the CHCl3 fraction CL-I (850 mg) and EtOAc fraction EA-I (330 mg). The pH of the residual aqueous layer was then adjusted to 8 with diluted NH3. It was then partitioned with CHCl3 (5 x 50 mL) and EtOAc (5 x 50 mL). Solvents were removed under vacuum, yielding the CHCl3 fraction CL-II (250 mg) and EtOAc fraction EA-II (133 mg). The pH of the aqueous layer was then adjusted to 7.0, and the solvent was removed by freeze-drying, giving fraction AQ (3.3 g). TLCs of the fractions CL-II and EA-II in several solvents (toluene/diethylamine/EtOAc 80:5:20; v/v CHCl3/MeOH/NH3 85:15:0.5; v/v) showed spots that gave a positive reaction to Dragendorf reagent. Fraction CL-II was repeatedly subjected to preparative thin layer chromatography (PTLC) with (toluene/diethylamine/EtOAc 80:5:20 v/v, CHCl3/MeOH/NH3 85:15:0.5 v/v). as eluents, yielding compounds 1 (10.1 mg) and 2 (6.5 mg). Fraction EA-II was subjected to silica plate chromatography using chloroform/methanol 4:1 as eluent. Further purification was achieved by semi-preparative HPLC with aqueous 0.1% HCOOH (A) and AcCN (B) as eluents. Gradient elution was used from 90% (A) to 85% (A) in 4 min, and then to 40% (A) in 22 min, yielding compound 3 (5.2 mg). The AQ fraction was subjected to several semipreparative HPLC steps (Spherisorb C-18, 1 x 300 mm, 10 μm) with aqueous 0.1% HCOOH (A) and methanol (B) as eluents. Gradient elution used for the isolation of compounds 4-7 was as follows: from 90% (A) to 85% (A) in 10 min, and then to 70% (A) in 15 min, yielding compounds 4 (11.0 mg) and 5 (13.0 mg); from 75% (A) to 45 % A in 25 min for the isolation of compounds 6 (8.0 mg) and 7 (6.5 mg). Navirine B (1) amorphous solid. [α]D: +12.0 (c 0.083, CH3OH). IR (KBr): 3350, 3030, 3020, 1670, 1650, 1610, 1508, 820 cm-1. 1 H NMR (300 MHz, CDCl3): 1.65-1.78 m (H-1), 1.30 m (H-2), 1.75-1.98 m (H-3), 1.87-2.02 m (H-6), 1.261.58 m (H-7), 1.64 m (H-9), 1.42-1.88 m (H-11), 2.54 m (H-12), 1.56-1.91 m (H-13), 5.67 brs (H-15), 4.55 brs (H-17), 1.05 s (H-18), 7.43 brs (H-19), 3.57 brs Dall’Acqua et al. (H-20), 6.85 d (8.0) (H-2’/6’), 7.12 d (8.0) (H-3’/5’), 2.99 brt (H-7’), 3.02 brt (H-8’), 2.40 s N-(CH3)2-9’. 13 C NMR (75.03 MHz, CDCl3): 27.9 (C-1), 30.1 (C-2), 30.9 (C-3), 44.9 (C-4), 72.6 (C-5), 30.9 (C-6), 20.9 (C-7), 43.8 (C-8), 47.1 (C-9), 41.0 (C-10), 43.2 (C-11), 31.7 (C-12), 43.5 (C-13), 69.8 (C-14), 130.8 (C-15), 146.8 (C-16), 68.8 (C-17), 19.1 (C-18), 169.9 (C-19), 80.6 (C-20), 158.2 (C-1’), 114.9 (C-2’/6’), 129.2 (C-3’/5’), 125.0 (C-4’), 33.3 (C-7’), 52.5 (C-8’), 34.3 (N-(CH3)2-9’). HRAPITOFMS: m/z [M + H+] calcd for C30H40N2O3: 477.3117; found: 477.3122. Navirine C (2) amorphous solid. [α]D: +7.5 (c 0.070, CH3OH). IR (KBr): 3350, 3030, 3018, 1650, 1605, 1508, 820 cm-1. 1 H NMR (300 MHz, CDCl3): 1.74-2.08 m (H-1), 1.62 m (H-2), 1.25-1.78 m (H-3), 1.26 m (H-5), 1.83-2.14 m (H-6), 1.80 m (H-7), 1.34-1.68 m (H-11), 2.87 m (H-12), 1.33 m (H-13), 5.63 brs (H-15), 4.52 brs (H-17), 1.00 s (H-18), 2.30 m (H-19), 2.53 m (H-20), 6.84 d (8.0) (H-2’/6’), 7.10 d (8.0) (H-3’/5’), 2.89 m (H-7’), 2.89 m (H-8’), 2.55 s (N-(CH3)2-9’). 13 C NMR (75.03 MHz, CDCl3): 21.8 (C-1), 28.5 (C-2), 34.1 (C-3), 41.7 (C-4), 53.5 (C-5), 33.9 (C-6), 34.1 (C-7), 44.4 (C-8), 44.5 (C-9), 44.0 (C-10), 28.1 (C-11), 31.1 (C-12), 44.1 (C-13), 45.0 (C-14), 132.4 (C-15), 145.8 (C-16), 68.7 (C-17), 27.8 (C-18), 58.0 (C-19), 76.7 (C-20), 158.8 (C-1’), 115.1 (C-2’/6’), 129.7 (C-3’/5’), 128.0 (C-4’), 33.2 (C-7’), 60.5 (C-8’), 44.3 (N-(CH3)2-9’). HRAPITOFMS: m/z [M + H+] calcd for C31H44N2O: 461.3532; found: 461.3512. Chellespontine (3) amorphous solid. [α]20D: +5.5 (c 0.052, CHCl3). IR (KBr) νmax: 2915, 1730, 990, 892 cm-1. 1 H NMR (300 MHz, CDCl3): 1.68 m (H-1), 1.31 m (H-2), 1.48-1.74 m (H-3), 1.38 m (H-5), 1.70 m (H-6), 1.61-2.05 (H-7), 2.19 m (H-9), 1.16-1.93 m (H-11), 2.40 m (H-12), 1.80-1.97 m (H-13), 1.301.93 m (H-14), 3.70 m (H-15), 5.05-5.12 d (J = 1.0) (H-17), 1.06 s (H-18), 3.77 m (H-19), 4.00 m (H-20), 3.75 m (H-21), 8.71 s (H-22). 13 C NMR (75.03 MHz, CDCl3): 25.7 (C-1), 19.6 ( C2), 40.5 (C-3), 33.5 (C-4), 45.6 (C-5), 19.7 (C-6), 34.2 (C-7), 37.2 (C-8), 39.8 (C-9), 46.0 (C-10), 27.9 (C-11), 36.1 (C-12), 25.3 (C-13), 27.9 (C-14), 74.7 Diterpenoid alkaloids and glycosides from Aconitum naviculare Natural Product Communications Vol. 3 (12) 2008 1989 (C-15), 154.8 (C-16), 109.7 (C-17), 24.0 (C-18), 59.5 (C-19), 56.5 (C-20), 65.0 (C-21), 183.3 (C-22). HRAPITOFMS: m/z [M + H+] calcd. for C22H33NO2: 344.2590; found: 344.2550. Cell growth was determined by the MTT reduction assay [12] after 72 h of incubation. Briefly, 20 μL of MTT solution (5 mg/mL in PBS) was added to each well and plates were incubated for 4 h at 37°C. DMSO (150 μL) was added to all wells and mixed thoroughly to dissolve the dark blue crystals. Absorbance was measured on a micro-culture plate reader (Titertek Multiscan) at a test wavelength of 570 nm and a reference wavelength of 630 nm. Experiments were performed at least in triplicate, and results were statistically evaluated using Student's t-test [13]. IC50 95% confidence limits were estimated with the Litchfield and Wilcoxon method. Compounds 4-7 were characterized on the basis of reported data [9-11]. Antiproliferative activity: Human ovarian carcinoma (2008) and human intestinal adenocarcinoma(LoVo) cell lines were used. The 2008 cells were maintained in RPMI 1640, and LoVo cells in Ham’s F 12, in both cases supplemented with 10% heat-inactivated FCS, 1% antibiotics and 1% 200 mM L-glutamine (all products of Biochrom KG Seromed). Cells were seeded on 96-well tissue plates (Falcon) at 5 x 104 cells/mL, and treated 24 h later with various concentrations of the compounds 1-3. Acknowledgment - The authors are grateful to MIUR for financial support. References [1] Amiya T, Bando H. (1988) Aconitum alkaloid, in The Alkaloids: Chemistry and Biology, Brossi A. (Ed) Academic Press Inc, San Diego, CA, Vol. 34, Chapter 3, 95-177. [2] Ameri A. (1998) The effects of Aconitum alkaloids on the central nervous system. Progress in Neurobiology, 56, 211-235. [3] Wada K, Hazawa M, Takahashi K, Mori T, Kawahara N, Kashiwakura I. (2007) Inhibitory effects of diterpenoid alkaloids on the growth of A172 human malignant cells. Journal of Natural Products, 70, 1854-1858. [4] Gao L, Wei X, Yang L. (2004) A new diterpenoid alkaloid from a Tibetan medicinal herb Aconitum naviculare Stapf. Journal of Chemical Research, 307-308. [5] Shrestha BB, Jha PK, Gewali MB. (2007) Ethnomedicinal use and distribution of Aconitum naviculare (Bruhl) Stapf in upper Manang, central Nepal. In: Local Effects of Global Changes in the Himalayas: Manang, Nepal. Chaudhary RP, Aase TH, Vetaas OR, Subedi BP. (Eds.) Tribhuvan University, Nepal, and University of Bergen, Norway, 171-181. [6] Shrestha BB, Dall’Acqua S, Gewali MB, Jha PK, Innocenti G. (2006) New flavonoid glycosides from Aconitum naviculare (Bruhl) Stapf, a medicinal herb from the trans-Himalayan region of Nepal. Carbohydrate Research, 341, 2161-2165. [7] Pelletier SW, Keith LH. (1970) In The Alkaloids: Chemistry and Physiology, Manske RHF. (Ed.) Academic Press, New York, Vol. 12, Chapter 2, 143-155. [8] Desai HK, Joshi BS, Pelletier SW, Sener B, Bingol F, Baykal T. (1993) New alkaloids from Consolida hellespontica. Heterocycles, 36, 1081-1089. [9] Du M, Xie J. (1995) Flavonol glycosides from Rhodiola crenulata, Phytochemistry, 38, 809-810. [10] Pauli GF. (2000) Higher order and substituent chemical shift effects in the proton NMR of glycosides. Journal of Natural Products, 63, 834-838. [11] Galland S, Mora N, Abert-Vian M, Rakotomanomana N, Dangles O. (2007) Chemical synthesis of hydroxycinnamic acid glucosides and evaluation of their ability to stabilize natural colors via anthocyanin copigmentation, Journal of Agricultural and Food Chemistry, 55, 7573-7579. [12] Mosmann T. (1983) Rapid colorimetric assay for cellular growth survival: application to proliferation and cytotoxic assay. Journal of Immunoogical Methods, 65, 55-63. [13] Tallarida RJ, Murray RB. (1987) Manual of Pharmacological Calculations with Computer Programs. Springer-Verlag. NPC Natural Product Communications Inhibition of PGHS-1 and PGHS-2 by Triterpenoid Acids from Chaenomelis fructus 2008 Vol. 3 No. 12 1991 - 1994 Eveline Reiningera and Rudolf Bauerb,* a Institute of Pharmaceutical Biology, Heinrich-Heine University, 40225 Düsseldorf, Germany b Institute of Pharmaceutical Sciences, Department of Pharmacognosy, Karl-Franzens-University, Universitätsplatz 4, 8010 Graz, Austria rudolf.bauer@uni-graz.at Received: July 14th, 2008; Accepted: October 28th, 2008 The dichloromethane extract of the dried fruits of Chaenomeles speciosa (Sweet) Nakai (Rosaceae) showed strong inhibitory activity against both prostaglandin-H-synthase isoenzymes [IC50 (PGHS-1) = 5.1 µg/mL; IC50 (PGHS-2) = 2.3 µg/mL]. The lipophilic portion of the extract was mainly responsible for the inhibitory effect. Several triterpenoid acids were isolated and identified as contributing to this inhibitory activity (oleanolic, pomolic, 3β-O-acetylursolic and 3β-O-acetylpomolic acids). Comparison of their inhibitory potential with their selectivity to PGHS-2 showed that 3β-O-acetylursolic acid had the highest potency in the inhibition of PGHS-1 and PGHS-2 enzymes, whilst pomolic and 3β-O-acetylpomolic acid, with a hydroxyl group at position 19α, showed selectivity for PGHS-2. The inhibitory effect of the extract seems to be the result of the activity of the mixture of these different triterpenoid acids. Keywords: Chaenomeles speciosa, Rosaceae, anti-inflammatory activity, prostaglandin-H-synthase, PGHS-1, PGHS-2, triterpenoid acids. Prostaglandin-H-synthase (PGHS) catalyses the first two steps of the formation of prostaglandins (PG) from arachidonic acid, with a fatty acid cyclooxygenase activity (catalyzing the reaction from arachidonic acid to PGG2) and a PG hydroperoxidase activity (catalyzing the reaction from PGG2 to PGH2). The enzyme exists in two isoforms: the constitutive PGHS-1, which is responsible for ‘housekeeping’ purposes, like the formation of protective PGs in the stomach and the kidneys, while PGHS-2 is mainly induced at sites of inflammation to form inflammatory PGs, mainly PGE2 [1]. PGE2 is a common marker for the determination of inflammation [2]. Therefore, PGHS is an important target of anti-inflammatory drug research. While common non-steroidal anti-inflammatory drugs act as unspecific inhibitors of both isoenzymes, the selective inhibition of PGHS-2 is the goal of new NSAIDs. They are expected to have less side effects on the stomach and the kidneys [3], even if it is known that long-term treatment with COX-2selective drugs (rofecoxib) for cancer prevention suggested an elevated incidence of myocardial infarction after long term use [4]. The dried fruits of Chaenomeles speciosa (Sweet) Nakai (“Mugua”), Rosaceae, are used as an antiinflammatory and antirheumatic drug in traditional Chinese medicine. The dichloromethane-extract of Mugua was tested for its inhibitory potential on prostaglandin synthesis in PGHS-1/-2 screening assays. We now report on the bioassay-guided fractionation and the identification of the PGHSinhibitory active principles. The fruits of C. speciosa were powdered and extracted with dichloromethane. The extract, when tested in PGHS-1 and -2 microtiter assays with EIA evaluation [5], showed very strong inhibition of both isoenzymes. The IC50 values were determined to be 5.1 µg/mL for PGHS-1 and 2.3 µg/mL for PGHS-2. The extract showed slight preferential inhibition of PGHS-2. When fractionated by vacuum liquid chromatography (VLC) with different mixtures of n-hexane/ethylacetate of increasing polarity there was a clear preference of inhibitory potential by the lipophilic part of the extract (Figure 1). 1992 Natural Product Communications Vol. 3 (12) 2008 Reininger et al. Table 1: PGHS-1/ -2 inhibition of triterpenoid acids from Chaenomelis fructus. 100 80 Conc. Inhibition [%] 60 Compounds Oleanolic acid Pomolic acid Maslinic acid Pirolonic acid Euscaphic/Tormentolic acid 40 20 0 I II III IV V VI VII VIII IX X -20 3β-O-Acetylursolic acid 3β-O-Acetylpomolic acid -40 -60 Figure 1: Inhibition (%) of PGHS-1/-2 in vitro by fractions from VLCseparation of the dichloromethane extract of Mugua (tested conc.: 50 µg/mL) HO HO COOH O Euscaphic acid HO HO HO 44.9 ± 8.8 48.2 ± 18.4 63.1 ± 6.9 56.7 ± 12.3 Table 2: IC50 values of the positive controls and the major triterpenoid acids of Mugua. Compound HO 100 97 * means from enriched fractions Fractions HO 110 106 106 103 102 Inhibition PGHS-1 PGHS-2 23.8 ± 27.7 10.4 ± 8.4 51.6 ± 8.2 49.4 ± 8.4 14.3 ± 7.4 11.8 ± 3.7 47.4 ± 4.1 43.4/21.4* 82.7/55.4* [µM] COOH Pirolonic acid Oleanolic acid Pomolic acid 3β-O-Acetylursolic acid 3β-O-Acetylpomolic acid Indometacin NS-398 IC50 PGHS-1 [µM] IC50 PGHS-2 [µM] ratio PGHS-1/ PGHS-2 204 68 34 165 0.9 50.7 348 36 39 90 0.8 2.6 0.6 2.0 0.9 1.8 1.1 19.5 COOH 3β-O-Acetylpomolic acid 3β-O-Acetylursolic acid HO HO Tormentolic acid From this part we isolated and identified different triterpenoid acids as the major constituents. The structures have been established on the basis of their spectral data (IR, EI-MS, DCI-MS, 1H NMR, 13C NMR) and by comparison with literature data [6-15] as 3β-O-acetylursolic acid, 3β-O-acetylpomolic acid, oleanolic acid, pomolic acid, euscaphic acid, tormentolic acid and pirolonic acid. The acids, reported for the first time from C. speciosa, were tested for their inhibitory activity of PGHSisoenzymes (Table 1). Most exhibited inhibition of both enzymes. Only euscaphic acid showed high inhibitory potential for PGHS-2. Separation of euscaphic and tormentolic acid was not achieved, so their inhibition values are derived from enriched fractions (ca. 90% purity). The IC50 values of the major triterpenoid acids are listed in Table 2. 3β-O-acetylursolic acid turned out to be the most potent inhibitor, while oleanolic acid showed only weak inhibition of both isoenzymes. When calculating the inhibition ratio PGHS-1/PGHS-2, pomolic acid and 3β-O-acetylpomolic acid showed more pronounced inhibitory activity against PGHS-2 than on PGHS-1. Both constituents possess a hydroxyl moiety in ring E. Figure 2 illustrates the selectivity by comparison of the ratios of PGHS1/PGHS-2 inhibition. Ratios > 1 are considered as preferential PGHS-1 inhibitors, ratio < 1 as preferential PGHS-2 inhibitors, a ratio = 1 as nonselective inhibitors. COOH COOH AcO AcO HO COOH COOH HO Pomolic acid Oleanolic acid HO PGHS-2 selective PGHS-1 3 2 1 2 3 Figure 2: Selectivities of the main triterpenoid acids of Mugua by comparison of the ratios of IC50 values (the ratios are calculated as PGHS-1/PGS-2 when preferential for PGHS-1, and PGHS-2/PGHS-1 when preferential for PGHS-2). Oleanolic and ursolic acid have already been described as potent inhibitors of PGHS [16,17]. In other test systems using microsomal preparations for PGHS-1 vs. pure enzyme for PGHS-2 [18], oleanolic and ursolic acid exhibited selectivity for PGHS-2. However, in the series of triterpenoid acids isolated from Mugua, oleanolic acid did not show the best inhibitory effect against either pure PGHS-1 or PGHS-2. Oleanolic and 3β-O-acetylursolic acid were more inhibitory on PGHS-1 than on PGHS-2. The selectivity was dependent on the 19α−hydroxyl moiety, found in pomolic acid and its derivatives. This observation could be confirmed by all isolated triterpenoid acids: all 19α-hydroxylated acids showed better inhibitory potential towards PGHS-2 than towards PGHS-1. A second hydroxyl moiety in ring A seems to intensify the preference. Triterpenoids as prostaglandin-H-synthase inhibitors Natural Product Communications Vol. 3 (12) 2008 1993 For high inhibitory potential, the α-amyrine-structure seems to be especially important. 3β-O-Acetylursolic acid turned out to be the most potent inhibitor. Acetylation at position 3β did not seem to influence the inhibitory potential. dissolved in EtOH p.a. for pharmacological testing at a concentration of 1 mg/mL. Interestingly, it was shown recently that oleanolic acid induced up-regulation of COX-2 via early phosphorylation of p38 MAPK and p42/44 MAPK [19], as was also found for alkamides of Echinacea and for the selective COX-2 inhibitor NS-398 in high concentration [20]. Oleanolic acid also induces rabbit platelet aggregation through a phospholipase C-calcium dependent signaling pathway [21]. Therefore, oleanolic acid may be considered as a modulatory compound for inflammation. Since about 70% of the dichloromethane extract of Mugua consists of triterpenoid acids, the very good inhibitory effect of the extract seems to be the result of the activity of these different acids. It will be interesting to demonstrate whether the inhibitory potential of triterpenoid acids can be further increased by structural modification. Experimental Extraction and fractionation: The plant material was provided by the TCM Hospital, Kötzting, Germany, and was identified in the Institute of Pharmaceutical Biology, Munich. A voucher specimen (12/1998) has been deposited there. The dichloromethane extract (17.5 g) was prepared by exhaustive Soxhlet extraction of 925 g powdered Chaenomelis fruits. This extract was fractionated in two portions by vacuum liquid chromatography (VLC) with n-hexane/ethyl acetate mixtures on 180 g Silica gel (Kieselgel 60, Merck) into fractions I - X. The fractions were evaporated to dryness and Table 3: Fractions obtained by VLC from the dichloromethane extract of Mugua. Fraction I II III IV V VI VII VIII IX X Eluent: n-Hexane/Ethyl acetate [mL] 300 : 0 280 : 20 270 : 30 250 : 50 230 : 70 210 : 90 190 : 110 170 : 130 150 : 150 130 : 170 120 : 180 90 : 210 60 : 240 30 : 270 0 : 300 Yield [g] 1.22 0.20 1.24 1.23 1.44 1.57 1.32 3.39 1.49 1.09 Isolation and structure elucidation of tested compounds: VLC fraction VI was further separated on Sephadex LH20 (100 g; column 90 x 2 cm ID) with a mixture of cyclohexane and ethyl acetate (6:4) as eluent, which resulted in a fraction of 550 mg phytosterols (sitosterol, campesterol) and ca. 55 mg triterpenoid acids. The purification of 3β-O-acetylpomolic acid (27.1 mg) and 3β-O-acetylursolic acid (28.0 mg) was achieved by MPLC on RP-18 (23 g LiChroprep RP 18, 25 – 40 µm, Merck; column 47 x 1 cm ID) with a water / acetonitrile gradient (10 – 40% in 10 min, 40 – 100% in 40 min). Fraction IX and X were first separated on Sephadex LH20 (100 g; column 90 x 2 cm ID) with ethyl acetate. Fraction IX was further separated by MPLC on RP-18 (8 g LiChroprep RP 18, Merck, 15 - 25 µm; column 40 x 0.8 cm ID) with a water/acetonitrile gradient (30 – 65% in 16 min, 65 – 90% in 34 min, 90 – 100% in 10 min). The final purification of oleanolic, pomolic and pirolonic acid was achieved by semipreparative HPLC on RP-18 (Hibar RT 250 x 10 mm ID, LiChrosorb RP 18, 7 µm, Merck) with a water/acetonitrile gradient (30 – 65% in 12 min, 65 – 78% in 14 min, 78 – 98% in 4 min, 98% 10 min). The identity of the compounds was confirmed on the basis of IR, DCI- and EI-MS, 1H and 13C NMR spectra in comparison with literature data [6-15]. The purity was checked by TLC and HPLC. Pharmacological assays: The PGHS-1 and PGHS-2 assays were performed on a microtiter scale with purified PGHS-1 from ram seminal vesicles and purified PGHS-2 from sheep placental cotyledones (both Cayman Chemical Company), as previously described [5]. The incubation mixture contained 180 µL 0.1 M tris-buffer (pH 8.0), 5 µM hematin, 18 mM epinephrin-hydrogentartrate, 0.2 U of enzyme preparation and 50 µM Na2EDTA (only PGHS-2 assay). Each compound solution (10 µL) was added and pre-incubated for 5 min at room temperature. The reaction was started by adding 10 µL of 5 µM arachidonic acid in EtOH p.a. and subsequent incubation at 37°C. The reaction was terminated after 20 min by adding 10 µL formic acid 10%. Determination of PGE2: The concentration of PGE2, the main metabolite of arachidonic acid in this reaction, was determined by a competitive PGE2EIA-kit (R&D Systems), which was used as described by Reininger et al. [5]. Because of the use 1994 Natural Product Communications Vol. 3 (12) 2008 of alkaline phosphatase, the procedure was shortened to 2 h incubation time and 45 min for the development process, which involves pNPP (p-nitrophenyl phosphate) as a substrate. The development was stopped by adding 2N NaOH. All samples were diluted 1:15 in EIA-buffer. The EIA was evaluated by an ELISA reader ‘rainbow’ (Tecan Deutschland GmbH, Crailsheim, Germany) and determined as previously described [5]. Inhibition refers to reduction of PGE2 formation in comparison with a blank without inhibitor. NS-398 and indomethacin (both from Cayman Chemical Company) were used as positive controls. Reininger et al. For testing, they were dissolved in EtOH p.a. at a concentration of 1 mM and further diluted with EtOH p.a.. Statistics: All IC50 values were determined for both enzymes by measuring at least three concentrations; all inhibition values are means of at least three experiments. Acknowledgments - We are grateful to the TCM Hospital, Kötzting, Germany, for supplying plant materials and for financial support, and we thank Mr Jansen for technical assistance. References [1] [2] [3] [4] [5] [6] [7] [8] [9] [10] [11] [12] [13] [14] [15] [16] [17] [18] [19] [20] [21] Hinz B, Brune K. (2002) Cyclooxygenase-2 – 10 years later, Journal of Pharmacology and Experimental Therapeutics, 300, 367-375. Vane JR, Botting RM. (1998) Anti-inflammatory drugs and their mechanism of action. Inflammation Research, 47 (Suppl. 2), S78-S87. Vane JT, Botting RM. (1995) New insights into the mode of action of anti-inflammatory drugs. Inflammation Research, 44, 1-10. Simmons DL, Botting RM, Hla T. (2004) Cyclooxygenase isozymes: the biology of prostaglandin synthesis and inhibition. Pharmacological Reviews, 56, 387-437. Reininger EA, Bauer R. (2006) Prostaglandin-H-synthase (PGHS)-1 and -2 microtiter assays for the testing of herbal drugs and in vitro inhibition of PGHS-isoenzyms by polyunsaturated fatty acids from Platycodi radix. Phytomedicine, 13, 164-169. Guo X, Zhang L, Quan S, Hong Y, Sun L, Liu M. (1998) Isolation and identification of triterpenoid compounds in fruit of Chaenomeles lagenaria (Loisel.) Koidz.. Zhungguo Zhongyao Zazhi, 23, 546-547. Mahato SB, Kundu AP. (1994) 13C NMR spectra of pentacyclic triterpenoids – a compilation and more important features. Phytochemistry, 36, 1517-1575. Kakuno T, Yoshikawa K, Arihara S. (1992) Triterpene saponins from fruit of Ilex crenata. Phytochemistry, 31, 2809-2812. Santos GG, Alves JCN, Rodilla JML, Duarte AP, Lithgow AM, Urones JG. (1997) Terpenoids and other constituents of Eucalyptus globulus. Phytochemistry, 44, 1309-1312. Yamaguchi K. (1970) Spectral Data of Natural Products, Vol. 1, Elsevier PC, Tokyo. Guang-YL, Gray A, Waterman PG. (1989) Pentacyclic triterpenes from fruits of Rosa sterilis. Journal of Natural Products, 52, 162-166. Takahashi K, Kawaguchi S, Nishimura KI, Kubota K, Tanabe Y, Takani M. (1974) Studies on the constituents of medical plants: XIII. Constituents of the pericarps of the capsules of Euscaphis japonica Pax. (1). Chemical & Pharmaceutical Bulletin, 22, 650-653. De Tommasi N, Rastrelli L, Cumanda J, Speranza G, Pizza C. (1996) Aryl and triterpenic glycosides from Margyriecarpus setosus. Phytochemistry, 42, 163-167. Brieskorn CH, Süss HP. (1974) Triterpenoide der Birnen- und Apfelschale. Archiv der Pharmazie, 307, 949-961. Yagi A, Okamura N, Haraguchi Y, Noda K, Nishioka I. (1978) Studies on the constituents of Zizyphi Fructus. II. Structure of new p-coumaroylates of maslinic acid. Chemical & Pharmaceutical Bulletin, 26, 3075-3079. Liu J. (1995) Pharmacology of oleanolic acid and ursolic acid. Journal of Ethnopharmacology, 49, 57-68. Díaz AM, Abad MJ, Fernández L, Recuero C, Villaescusa L, Silván AM, Bermejo P. (2000) In vitro anti-inflammatory activity of iridoids and triterpenoid compounds isolated from Phillyrea latifolia L. Biological & Pharmaceutical Bulletin, 23, 1307-1313. Ringbom T, Segura L, Noreen Y, Perera P, Bohlin L. (1998) Ursolic acid from Plantago major, a selective inhibitor of cyclooxygenase-2 catalyzed prostaglandin biosynthesis. Journal of Natural Products, 61, 1212-1215. Martínez-González J, Rodríguez-Rodríguez R, González-Díez M, Rodríguez C, Herrera MD, Ruiz-Gutierrez V, Badimon L. (2008) Oleanolic acid induces prostacyclin release in human vascular smooth muscle cells through a cyclooxygenase-2-dependent mechanism. Journal of Nutrition, 138, 443-448. Hinz B, Woelkart K, Bauer R. (2007) Alkamides from Echinacea inhibit cyclooxygenase-2 activity in human neuroglioma cells. Biochemical and Biophysical Research Communications, 360, 441-446. Lee JJ, Jin YR, Lim Y, Yu JY, Kim TJ, Yoo HS, Shin HS, Yun YP. (2007) Oleanolic acid, a pentacyclic triterpenoid, induces rabbit platelet aggregation through a phospholipase C-calcium dependent signaling pathway. Archives of Pharmacal Research, 30, 210-214. NPC 2008 Vol. 3 No. 12 1995 - 1997 Natural Product Communications Preparative Isolation of Antimycobacterial Shoreic Acid from Cabralea canjerana by High Speed Countercurrent Chromatography Gilda G. Leitãoa*, Lisandra F. Abreua, Fernanda N. Costaa, Thiago B. Bruma, Daniela Fernandes Ramosb, Pedro Eduardo A. Silvab, Maria Cristina S. Lourençoc and Suzana G. Leitãod a Núcleo de Pesquisas de Produtos Naturais, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Bl. H, CCS. Ilha do Fundão, Rio de Janeiro, RJ, Brazil, 21.941-590 b Universidade Federal do Rio Grande, FURG, Departamento de Patologia, Laboratório de Micobactérias, Rua General Osório S/N Área Acadêmica da Saúde, CEP: 96200-190 Rio Grande/RS, Brazil c Instituto de Pesquisa Clínica Evandro Chagas, Fiocruz Laboratório de Bacteriologia e Bioensaios em Micobactérias, Plataforma de Bioensaios II, FIOCRUZ, 21045-900, Rio de Janeiro, Brazil d Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Bl. A, CCS. Ilha do Fundão, Rio de Janeiro, RJ, Brazil, 21.941-590 ggleitao@nppn.ufrj.br Received: July 14th, 2008; Accepted: November 11th, 2008 High speed countercurrent chromatography (HSCCC) was used to isolate the dammarane type triterpene shoreic acid from the dichloromethane extract of leaves of Cabralea canjerana, which showed activity against Mycobacterium tuberculosis. A preparative scale-up of the process was also developed. Keywords: High-speed countercurrent chromatography, HSCCC, Cabralea canjerana, tuberculosis, antimycobacterial activity, Meliaceae. The separation of bioactive secondary metabolites from crude plant extracts has always been a challenge to natural products researchers and countercurrent chromatography (CCC) offers many advantages compared with traditional phytochemical techniques of purification, especially those where chromatography with solid supports is used. The main advantage of CCC is that it is a form of liquid-liquid chromatography, which does not use a solid support and, therefore, there can be no loss of compounds or bioactivity due to interactions between the solid phase and the target compounds [1]. In the course of our investigation of bioactive plants from the Brazilian Atlantic forest, the dichloromethane extract of leaves of Cabralea canjerana (Meliaceae) showed antimycobacterial activity (minimal inhibitory activity, MIC of OH H O HO2C Figure 1: Shoreic Acid. 100 μg/mL) and was fractionated by high-speed countercurrent chromatography (HSCCC). The success of any HSCCC fractionation is dependent on the correct choice of solvent system, as this form of chromatography is based on the partition of solutes 1996 Natural Product Communications Vol. 3 (12) 2008 Table 1: Amount of dichloromethane extract injected into the HSCCC equipment (sample size) and corresponding amount of shoreic acid obtained in the isolation process. Solvent system n-hexane:EtOAc:MeOH:H2O 1:1.5:2.5:1. a Sample size (g) Fraction no.a Fraction size (mL) 0.8 1 1.5 17-25 14 -24 17-26 4 4 5 Shoreic acid (mg) 213 321 642 fractions where shoreic acid was eluted. between two immiscible liquid phases. Compounds are separated according to their distribution constants (KD), expressed as the ratio of their concentration in the stationary phase to their concentration in the mobile phase [2]. The test-tube partitioning test [3] is a good way of predicting the distribution ratios of target compounds to be separated and was used here for the choice of the appropriate solvent system. As the bioactive extract of C. canjerana contained medium polarity compounds, the solvent system nhexane-ethyl acetate-methanol-water was chosen. Various ratios of the solvents in the biphasic solvent system were tested: n-hexane-ethyl acetate-methanolwater 1:2:1:2, 1:1.5:2.5:1, 1:2:2:1; 1:2:2.5:0.5, 1:2:2.5:1, (v:v:v:v). The best solvent system is that when the KD of the target compounds remains around 1. Also, the volumes of upper and lower phases should be equivalent. When this is not the case (as in some of the solvent systems tested), the chosen system should be that where the mobile phase has a larger volume. Bearing this in mind, the final solvent system chosen for this fractionation was n-hexaneethyl acetate-methanol-water 1:1.5:2.5:1 (v:v:v:v), with the upper organic layer acting as the stationary phase and the lower aqueous layer as the mobile phase. In this mode of CCC fractionation (reversed phase mode), the more polar compounds in the extract elute first. The separation was initially run with 820 mg of the dichloromethane extract of leaves of C. canjerana, affording ca. 213 mg of a dammarane triterpene, shoreic acid (KD approx. 1) (Fig. 1), the major compound in this extract. The structure of this compound was confirmed by 1H and 13 C NMR spectroscopic data, which were in accordance with those in the literature [4]. The activity of this compound against Mycobacterium tuberculosis was tested by the MABA as well as the REMA assays, showing a MIC of 100 μg/mL. Less polar triterpenes were retained in the stationary organic phase, which was also fractionated, affording other triterpenes (structures under investigation). CCC is particularly useful in the preparative range (mg to grams) and the time required for preparative Leitão et al. separations is no more than a few hours. The previous separation took about 3 h and consumed about 1.5 L of solvent. The isolation of shoreic acid was scaled-up from 820 mg to 1.5 g, using the same column volume, with good reproducibility (Table 1). This method proved to be fast, economic and highly effective in the scaled-up isolation of shoreic acid directly from a crude plant extract. Experimental General procedures: The NMR spectra were recorded using a Bruker Avance DRX400 spectrometer (Karlsruhe, Germany) at 25oC, 1 operating at 400.13 MHz for H and 100.61MHz for 13 C. NMR spectra were recorded in CDCl3 using TMS as internal standard. TLC analyses were carried out on pre-coated silica gel plates GF254 from Merck, and visualized by UV (254 nm) and reaction with vanillin in sulfuric acid (2%), followed by heating. For HSCCC separations a P.C. Inc countercurrent chromatograph equipped with a triple multi-layer coil equilibrated by a counterweight was used. Solvents were pumped into the coil with a Rainin Dynamax Model SD-200 pump. A Rainin Dynamax FC-1 fraction collector was also used. Plant material and extraction: Leaves of Cabralea canjerana (Vell.) Mart. were collected in May 2003 at Mata Boa Vista Farm, Levy Gasparian, Rio de Janeiro State, Brazil. The plant was identified by Sebastião José Silva Neto, from the Instituto de Pesquisas Jardim Botânico do Rio de Janeiro, and a voucher specimen is deposited at the herbarium of the Federal University of Rio de Janeiro. The dried and ground leaves (890 g) were exhaustively extracted with ethanol 96oGL. The resulting dried ethanolic extract was suspended in water-methanol 70:30 (v:v) and extracted with n-hexane, dichloromethane, ethyl acetate and n-butanol, in this order. Choice of solvent system by test tube experiments: Small amounts of the dichloromethane extract from leaves of C. canjerana were dissolved in separate test tubes containing n-hexane-ethyl acetate-methanolwater 1:2:1:2, 1:2:2.5:0.5, 1:2:2:1; 1:2:2.5:1, and 1:1.5:2.5:1 (v:v:v:v). The test tubes were shaken and the compounds allowed to partition between the two phases. Equal aliquots of each phase were spotted beside each other separately on silica gel TLC plates and developed with CHCl3:MeOH 6:0.5 (v:v). The results were visualized by spraying with vanillin in sulfuric acid (2%), followed by heating. The final HSCCC isolation of shoreicacid from Cabralea canjerana Natural Product Communications Vol. 3 (12) 2008 1997 solvent system was set as n-hexane-ethyl acetatemethanol-water 1:1.5:2.5:1. Antimycobacterial tests: Samples were simultaneously screened by both microbiology laboratories (FURG and FIOCRUZ), using the MABA and REMA bioassays, respectively, as described previously [5]. Final concentration of plant extracts/substances was either 200 μg/mL or 100 μg/mL. Media plus bacteria with and without rifampicin were used as controls. The strain H37Rv (ATCC - 27294) was used for all methodologies. MABA (Microplate Alamar Blue Assay) susceptibility testing was performed at FIOCRUZ according to the method described in [6]. The REMA - Resazurin Microtiter Assay Plate [7] method was used for the determination of the antimycobacterial activity at FURG. In brief, the assay is accomplished in microplates (96 wells) using resazurin as indicator of cellular viability. Medium 7H9 enriched with 10% OADC was used. The minimal inhibitory concentration (MIC) was determined (starting at 200 µg/mL in 1:2 serial dilutions). HSCCC separation: The volume of the coil used in the experiments was 80 mL. The CCC column was filled with the organic stationary phase of the solvent system n-hexane-ethyl acetate-methanolwater 1:1.5:2.5:1. After the coil had been filled with the stationary phase, rotation started and the aqueous mobile phase was pumped into the head to tail direction at 2 mL/min until hydrodynamic equilibrium was achieved. In these conditions, the organic stationary phase, VS, initially retained in the CCC column was 67 mL (Sf = 75 %; VM = 20 mL). Eight hundred and twenty milligrams of the dichloromethane extract of leaves of C. canjerana was dissolved in 2.5 mL of each phase of the solvent system. The 5 mL was injected in the 80 mL coil using a Rheodyne injection valve at a flow rate of 2 mL/min., 850 rpm; 57 fractions of 4 mL were collected. Rotation stopped at tube 40 (which corresponds to KD = 2). Shoreic acid (213 mg) was obtained from fractions 17 – 25, corresponding to a KD of approximately 1. Further fractionations were carried out with 1 g and 1.5 g of the dichloromethane extract (Table 1). The same 80 mL coil was used, but the injection volumes were now 10 mL for both experiments. All other conditions were the same, except for the volume of fractions collected in the 1.5 g separation, which was 5 mL. Acknowledgments - The authors wish to thank CNPq (Edital MCT-CNPq/MS-SCTIE-DECIT 25/2006, Process no. 410475/2006-8), FAPERJ (E-26/170.442/03) for financial support. We are also indebted to Centro Nacional de Ressonância Magnética Nuclear Jiri Jones, UFRJ, Rio de Janeiro, for the use of NMR equipment. Collaborative work was performed under the auspices of the Iberoamerican Program for Science and Technology (CYTED), Project X.11:PIBATUB. References [1] Alvi KA. (2001) Screening natural products:bioassay-directed isolation of active components by dual mode CCC, Journal of Liquid Chromatography & Related Technologies, 24, 1765-1773. [2] Conway WD, Chadwick LR, Fong HHS, Farnsworth NR, Pauli GF. (2005) Extra column volume in CCC, Journal of Liquid Chromatography & Related Technologies, 28, 1799-1818. [3] Berthod A, Carda-Broch S. (2004) Determination of liquid-liquid partition coefficients by separation methods. Journal of Chromatography A, 1037, 3-14. [4] Roux D, Martin MT, Adeline MT, Sevenet T, Hadi H, Pais M. (1998) Foveolins A and B, dammaranes triterpenes from Aglaia foveolata. Phytochemistry, 49, 1745-1748. [5] Leitão SG, Castro O, Fonseca EN, Julião LS, Tavares ES, Leo RRRT, Vieira RC, Oliveira DR, Leitão GG, Martino V, Sülsen V, Barbosa YAG, Pinheiro DPG, Silva PEA, Teixeira DF, Neves-Junior I, Lourenço MCS (2006) Screening of Central and South American plant extracts for antimycobacterial activity by the Alamar Blue test. Brazilian Journal of Pharmacognosy, 16, 6-11. [6] Franzblau SG, Witzig RS, Mclaughlin JC, Torres P, Madico G, Hernandez A, Degnan MT, Cook MB, Quenzer VK, Ferguson RM, Gilman RH. (1998) Rapid, low-technology MIC determination with clinical Mycobacterium tuberculosis isolates by using the Microplate Alamar Blue Assay. Journal of Clinical Microbiology, 36, 362-366. [7] Palomino JC, Martin A, Camacho M, Guerra H, Swings J, Portaels F. (2002) Resazurin microtiter assay plate: Simple and inexpensive method for detection of drug resistance in Mycobacterium tuberculosis. Antimicrobial Agents and Chemotherapy, 46, 2720-2722. NPC Natural Product Communications Antiplasmodial Effects of a few Selected Natural Flavonoids and their Modulation of Artemisinin Activity 2008 Vol. 3 No. 12 1999 - 2002 Anna Rita Biliaa,*, Anna Rosa Sannellab, Franco Francesco Vincieria, Luigi Messoric, Angela Casinic, Chiara Gabbiani c, Carlo Severini b and Giancarlo Majori b a Department of Pharmaceutical Sciences, University of Florence, Via U. Schiff 6, 50019 Sesto Fiorentino, Florence, Italy b Department of Infectious, Parasitic and Immunomediated Diseases, Vector-Borne Diseases and International Health Section, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy c Department of Chemistry, University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Florence, Italy ar.bilia@unifi.it Received: May 16th, 2008; Accepted: July 26th, 2008 The direct antiplasmodial effects of five structurally-related flavonoids, namely quercetin, rutin, eriodictyol, eriodictyolchalcone and catechin, were analyzed in vitro on P. falciparum. Notably, all these flavonoids, with the only exception of rutin, caused relevant inhibition of P. falciparum growth when given at 1 mM concentration. In addition, they were found to affect greatly the potent antiplasmodial activity of artemisinin, leading to significant additive and even synergistic effects. In particular, quercetin induced a pronounced synergistic effect. The observed synergisms might be conveniently exploited to design new and/or more effective combination therapies. Keywords: Flavonoids, quercetin, rutin, eriodictyol, eriodictyolchalcone and catechin, artemisinin, Plasmodium falciparum, additive and synergistic effects. Artemisinin, a sesquiterpene endoperoxide isolated from Artemisia annua L., is today a leading drug for the treatment of malaria. It is very effective against severe forms of malaria and, in particular, against chloroquine-resistant, primaquine-resistant and some other life-threatening forms of malaria. It is a drug of high efficacy and rapid action, with a good tolerability, leading to clearance of parasites from the blood within two days [1]. Artemisinin and its semi-synthetic derivatives induce a very rapid reduction of parasitaemia, starting almost immediately after administration and are potent blood schizontocides [2]. These compounds are also gametocytocidal [3] and, interestingly, block the early sexual-stage (gametocyte) development [4,5], but they do not kill the hepatic stages of the parasite. In addition, they inhibit important pathophysiological processes, such as cytoadherence of Plasmodium falciparum-infected erythrocytes to microvasculature, more effectively antimalarial drug classes [6]. than other The mechanism of action of artemisinin and its derivatives is still a matter of intense debate [7,8]. Their activity is strictly associated to the presence of a unique structural feature, the endoperoxide bridge; their primary target, however, is yet not known. Meunier and coworkers demonstrated that heme is alkylated by artemisinin in meso positions and an extensive characterization of such adducts was carried out accordingly [9-11]. Heme alkylation was recently shown to occur also in vivo [12]. Similar heme adducts are formed in vitro with some derivatives of artemisinin, as found in our previous investigation [13]. A significant reactivity of artemisinin with hemoglobin has been described as well [14]. However, more recently, it was proposed that artemisinin and its derivatives most likely act through a mechanism similar to thapsigargin. This 2000 Natural Product Communications Vol. 3 (12) 2008 Bilia et al. latter drug potently inhibits PfATP6, a key parasite Ca2+ transporter with a particular selectivity for the SERCA of malarial PfATP6 rather than for mammalian pumps [15]. Within the frame of a larger research project aimed at evaluating the possible synergism/antagonism of antimalarial drugs with a variety of widespread natural products, we have recently shown that green tea extracts, as well as their main catechin constituents, markedly inhibit P. falciparum growth in vitro and potentiate the antimalarial effects of artemisinin [17]. These results are nicely consistent with the concept that complex mixtures of artemisinin with other natural products, as those that are usually found in the crude plant extracts, are often more effective than the purified drug itself. The higher antimalarial potency of the extract compared to the pure drug has thus been ascribed to the concomitant and independent biochemical actions of the other constituents as well as to the occurrence of appreciable synergistic interactions. A number of additional experimental and clinical observations support these ideas. In view of this background and of the known antimalarial properties of other flavonoids [18], this article describes the effects against P. falciparum 3D7 strain, sensitive to chloroquine, of a group of structurally-related flavonoids, namely the flavonol quercetin (2), its diglycoside rutin (3), the flavanone eriodictyol (4), the chalcone eriodictyolchalcone ((E)-3-(3,4-dihydroxy-phenyl)-1-(2,4,6-trihydroxyphenyl)-propenone) (5), and catechin (6). These flavonoids were assayed either alone or in combination with artemisinin. The first step of our investigation was the evaluation of the effects of the above flavonoids, administered alone, on P. falciparum growth, using two typical and well separated drug concentrations, namely 10 μM and 1 mM, after 48 hours incubation. Growth inhibition was evaluated by a well established 90 10 m icroM 1mM 80 70 % inhibition Due to the evident lack of conclusive ideas concerning the mechanism of action, together with the fact that artemisinin kills parasites rapidly, but is also rapidly excreted, the WHO Expert Consultative Group in 2001 strongly recommended the development of combination therapies since such drug associations have the potential to reduce the appearance of resistance and to be more effective in eradicating malaria [16]. 100 60 50 40 30 20 10 0 Rutin Quercetin Catechin Eriodictyol Eriodictyol chalcone Figure 1: Percentage inhibitions of 3D7 P. falciparum strains against two different concentrations (1mM and 10 μM) of flavonoids. The data are expressed assuming no inhibition of untreated controls. enzymatic method based on lactate dehydrogenase activity of P. falciparum (pLDH). Results are reported in Figure 1. From inspection of Figure 1, it is evident that all these flavonoids as such, produce rather modest growth inhibition of Plasmodium 3D7 strain, if given at 10 μM concentration. Indeed, only quercetin, eryodictiol and its chalcone produced some measurable effect, causing growth decreases of ~ 5-15% (see Figure 1). In contrast, large inhibitory effects were seen when flavonoids were given at 1 mM concentration. In this latter case, eryodictiol chalcone and catechin turned out to be the most active with 88% and 86% inhibition, respectively. Using the same 1 mM concentration, eryodictiol caused 68% inhibition and quercetin 51% inhibition. In contrast, rutin, the diglycoside of quercetin, had only a modest activity causing just 10% inhibition. It is well known that these substances are usually safe and that their concentrations in many plant extracts may be very high; this observation implies that even relatively moderate antimalarial activities, such as those reported above, may have, nonetheless, important practical consequences. Afterwards, possible synergism between artemisinin and the single flavonoid constituents 2-6 on parasite growth was investigated by monitoring artemisinin effects in the presence (or in the absence) of either 10 µM or 1 mM concentrations of the individual compounds. In this second set of experiments, ART concentration ranged between 0.625 to 40 nM, as administration of such concentrations resulted in partial inhibition of P. falciparum growth (Figure 2). Notably, we found that the effects of artemisinin, given at different concentrations, and 10 µM quercetin are more than additive; this effect becomes still more evident when quercetin was tested at 1 mM Antiplasmodial effects of flavonoids quercetin 1 artemisinin 100 90 In particular, the observed synergism between quercetin and artemisinin might be conveniently exploited to design new and/or more effective combination therapies. Moreover, we believe that valuable mechanistic information on the individual antimalarial agents may be extracted from a careful comparative analysis of the established positive and negative drug-drug interactions [20]. artemisinin plus quercetin 1 80 % inhibition Natural Product Communications Vol. 3 (12) 2008 2001 70 60 50 40 30 Experimental 20 10 0 0 0.625 1.25 2.5 5 10 20 40 concentration artemisinin (nM ) Figure 2: Percentage inhibition of 3D7 P. falciparum strain against quercetin 1 mM, increasing concentrations (from 0.625 nM to 40 nM) of artemisinin and increasing concentrations (from 0.625 nM to 40 nM) of artemisinin plus quercetin 1 mM. The data are expressed assuming no inhibition of untreated controls. concentration, implying some significant synergism between these two substances. The largest synergistic effects are detected for (sublethal) ART doses ranging from 0.625 to 10 nM (Figure 2). The other flavonoids did not show evident synergistic effects; however, sizable additive effects were found when eriodictyol and eriodictyolchalchone 1 mM were added to 5, 2.5, 1.25 and 0.625 nM of artemisinin. These flavonoids at 10 μM concentrations did not show additive effects. Overall, the results reported above point out that the intrinsic antiplasmodial activities of the five tested flavonoids are rather moderate or even modest. However, they cannot be neglected in view of the usually high concentrations of these substances in plant extracts and of their safety. In addition, we have shown that all these flavonoids generally manifest additive effects with artemisinin, leading to a substantial potentiation of its antimalarial actions. Most remarkably, considerable synergism was found in the case of quercetin. This latter finding is not surprising as quercetin is known to be an inhibitor of thioredoxin reductase and a prooxidant [19]. As artemisinin is thought to work through induction of oxidative stress on P. falciparum, the observed synergism might be straightforwardly ascribed to exacerbation of oxidative stress caused by quercetin. Reagents: Artemisinin was obtained from Sigma (Milan, Italy). Flavonoids 2-6 were from Extrasynthese (Genay, France). Parasite maintenance: Cultures of P. falciparum, 3D7 drug-sensitive to chloroquine (CQR), were grown in vitro in human red blood cells (O+), as formerly described [21]. In vitro determination of antimalarial activity of flavonoids: The selected flavonoids were prepared as stock solutions in 95% ethanol and then diluted in complete RPMI medium (containing 10% human serum). In all tests, the concentration of ethanol was maintained at 0.02% and did not inhibit the growth of control cultures. The individual flavonoids were presented at concentrations of 10 μM and 1 mM. Growth inhibition was evaluated by a well established enzymatic method based on lactate dehydrogenase activity of P. falciparum (pLDH) [22]. The parasite strain of P. falciparum, 3D7 drugsensitive to chloroquine, was used for the in vitro tests after culture syncronization by sorbitol [23] . The effects of flavonoids on cultured parasites were determined by light microscopy and pLDH activity, as previously described in detail [24]. Each assay was performed in triplicate, on 3 separate occasions. Monitoring flavonoid modulation of artemisinin activity: P. falciparum growth inhibition was analysed as a function of added artemisinin, at sublethal doses, ranging from 0.625 to 40 nM, either in the presence or absence of either 10 μM or 1 mM of the flavonoid constituents. Acknowledgments - The Ente Cassa di Risparmio di Firenze is gratefully acknowledged for generous financial support. References [1] Tu Y. (2004) The development of the antimalarial drugs with new type of chemical structure-qinghaosu and dihydroqinghaosu. Southeast Asian Journal of Tropical Medicine and Public Health, 35, 250-251. 2002 Natural Product Communications Vol. 3 (12) 2008 Bilia et al. [2] Balint GA. (2001) Artemisinin and its derivatives: an important new class of antimalarial agents. Pharmacology and Therapy, 90, 261-265. [3] Dhingra V, Vishweshwar Rao K, Lakshmi Narasu M. (2000) Current status of artemisinin and its derivatives as antimalarial drugs. Life Sciences, 66, 279-300. [4] Price RN, Nosten F, Luxemburger C, Kuile FO, Paiphun L, Chongsuphajaisiddhi T, White NJ. (1996) Effects of artemisinin derivatives on malaria transmissibility. Lancet, 347, 1654-1658. [5] Targett G, Drakeley C, Jawara M, von Seidlein L, Coleman R, Deen J, Pinder M, Doherty T, Sutherland C, Walraven G, Milligan P. (2001) Artesunate reduces but does not prevent posttreatment transmission of Plasmodium falciparum to Anopheles gambiae. Journal of Infections and Diseases, 183, 1254-1259. [6] Udomsangpetch R, Pipitaporn B, Krishna S, Angus B, Pukrittayakamee S, Bates I, Suputtamongkol Y, Kyle DE, White NJ. (1996) Antimalarial drugs reduce cytoadherence and rosetting Plasmodium falciparum. Journal of Infections and Diseases, 173, 691-698. [7] O'Neill PM, Posner GH. (2004) A medicinal chemistry perspective on artemisinin and related endoperoxides. Journal of Medicinal Chemistry, 47, 2945-2964. [8] Haynes RK., Krishna S. (2004) Artemisinins: activities and actions. Microbes and Infections, 6, 1339-1346. [9] Cazelles J., Robert A, Meunier B. (2002) Alkylating capacity and reaction products of antimalarial trioxanes after activation by a heme model. Journal of Organic Chemistry, 67, 609-619. [10] Robert A, Coppel Y, Meunier B. (2002) Alkylation of heme by the antimalarial drug artemisinin. Chemical Communications, 414-415. [11] Robert A, Dechy-Cabaret O, Cazelles J, Meunier B. (2002) From mechanistic studies on artemisinin derivatives to new modular antimalarial drugs.Accademy of Chemical Research, 35, 167-174. [12] Robert A, Benoit-Vical F, Claparols C, Meunier B. (2005) The antimalarial drug artemisinin alkylates heme in infected mice. Procedings of the National Academy of Sciences of the United States of America, 102, 13676-13680. [13] Messori L, Piccioli F, Eitler B, Bergonzi MC, Bilia AR, Vincieri FF. (2003) Spectrophotometric and ESI-MS/HPLC studies reveal a common mechanism for the reaction of various artemisinin analogues with hemin. Bioorganic and. Medicinal Chemistry Letters, 13, 4055-4057. [14] Messori L, Gabbiani C, Casini A, Siragusa M, Vincieri FF, Bilia AR. (2006) The reaction of artemisinins with hemoglobin: a unified picture. Bioorganic and. Medicinal Chemistry, 14, 2972-2977. [15] Eckstein-Ludwig U, Webb RJ, Van Goethem ID, East JM, Lee AG, Kimura M, O'Neill PM, Bray PG, Ward SA, Krishna S. (2003) Artemisinins target the SERCA of Plasmodium falciparum. Nature, 424, 887-889. [16] World Health Organization, 2001. Antimalarial Drug Combination Therapy: Report of a Technical Consultation. WHO, Geneva. [17] Sannella AR, Messori L, Casini A, Vincieri FF, Bilia AR, Majori G, Severini C. (2007) Antimalarial properties of green tea. Biochemical and Biophysical Research Communications, 353, 177-181. [18] Bilia AR. (2006) Non-nitrogenous plant-derived constituents with antiplasmodial activity. Natural Product Communications, 1, 1181-1204. [19] Lu J, Papp LV, Fang J, Rodriguez-Nieto S, Zhivotovsky B, Holmgren A. (2006) Inhibition of mammalian thioredoxin reductase by some flavonoids: implications for myricetin and quercetin anticancer activity. Cancer Research, 66, 4410-4418. [20] Bell A. (2005) Antimalarial drug synergism and antagonism: mechanistic and clinical significance. FEMS Microbiological Letters, 253, 171-184. [21] Trager W, Jensen JB. (1976) Human malaria parasites in continuous culture. Science, 93, 673-675. [22] Makler MT, Ries JM, Williams JA, Bancroft JE, Piper RC, Gibbins BL, Hinrichs DJ. (1993) Parasite lactate dehydrogenase as an assay for Plasmodium falciparum drug sensitivity. American Journal of Tropical Medicine and Hygiene, 48, 739-741. [23] Lambros C, Vanderberg JP. (1979) Synchronization of Plasmodium falciparum erythrocytic stages in culture. Journal of Parasitology, 65, 418-420. [24] Severini C, Menegon M, Sannella AR, Paglia MG, Narciso P, Matteelli A, Gulletta M, Caramello P, Canta F, Xayavong MV, Moura INS, Pieniazek NJ, Taramelli D, Majori G. (2006) Prevalence of pfcrt point mutations and level of cloroquine resistance in Plasmodium falciparum isolates from Africa, Infections and Genetic Evolution, 6, 262-268. NPC Natural Product Communications Comparative Analysis of Antimalarial Principles in Artemisia annua L. Herbal Drugs from East Africa 2008 Vol. 3 No. 12 2003 - 2006 Silvia Lapenna§, Maria Camilla Bergonzi, Franco Francesco Vincieri and Anna Rita Bilia* Department of Pharmaceutical Sciences, via Ugo Schiff,6, Universit of Florence, Sesto Fiorentino (FI), Italy 50019 § Present address: Centro di Ricerche Oncologiche “Fiorentino Lo Vuolo” (CROM), Via Ammiraglio Bianco, 83013, Mercogliano (AV), Italy ar.bilia@unifi.it Received: July 25th, 2008; Accepted: October 23rd, 2008 Malaria mortality continues to increase across the world and represents the most important parasitic disease of man. Artemisia annua L. (Asteraceae) has been used to treat fevers in China for over two millennia and recently the clinical efficacy of teas and decoctions derived from this species have been demonstrated, using high artemisinin-yielding plants. Therefore, it is important to verify the artemisinin levels in local cultivations in areas where malaria is endemic and to assess how different geographical and climatic conditions may affect the efficacy of traditional treatments. In this study, samples of the aerial parts of A. annua (ANAMED 3 hybrid) cultivated in three different locations in Burundi were compared for their content of active principles. Artemisinin levels in the plant materials ranged from 0.20% to 0.35%, while total flavonoid contents ranged from 0.32% to 0.80%. Keywords: Artemisia annua L., Burundi, different cultivations, artemisinin, flavonoids, HPLC/DAD/MS. Malaria is one of the oldest and most important lifethreatening parasitic diseases in the tropical regions of the world. It causes more than 300 million acute illnesses and at least 1-2.7 million deaths annually. The majority of these deaths are due to cerebral malaria and other complications resulting from malaria-related anaemia, and the cost in human life, incapacity for work, programs of control and medical treatments are enormous [1a-1c]. Ninety per cent of those who die are in Africa, where malaria accounts for about one in five of all childhood deaths, mainly children under the age of five in sub-Saharan Africa. Burundi is among the African countries with a high incidence of malaria, which is probably the leading cause of death in this as well as other East African countries [1d]. The 2000-2001 epidemic of Plasmodium falciparum in Burundi, with an attack rate peak of 109% in the northern provinces of Kayanza, Karuzi, and Ngozi, is well documented [1e,1f]. Some non-governmental organizations and international agencies working in Burundi have offered to introduce to these regions the use of the plant Artemisia annua L. (sweet or annual wormwood) because its active constituent, artemisinin, has proven suitable for the control of malaria epidemics, including chloroquine- and quinine-resistant strains, and has shown a low propensity to induce resistance [2]. A. annua is an annual herb native to the northern parts of Chahar and Suiyuan provinces in China, where it is called “quinghao” and has been used as a remedy for chills and fevers for more than 2000 years [3,4]. In the most recent literature, clinical trials using teas or decoctions of A. annua leaves from high artemisinin-yielding plants (> 0.5% dried weight) grown in Central Africa, have shown a rapid disappearance of malaria parasites from the blood of patients treated with doses corresponding to the Chinese Pharmacopoeial recommendations [5-7]. Artemisinin plasma concentrations after intake of these A. annua traditional preparations were lower 2004 Natural Product Communications Vol. 3 (12) 2008 than those achieved with modern artemisinin drugs used in malaria therapy, but still above 10 μg/L, the threshold for parasite growth inhibition. Therefore, the locally grown herb may offer an additional tool for the control of malaria, especially in poor countries with scarce or no access to modern medicines or medical services. However, it is known that levels of artemisinin in A. annua may vary considerably (0.01-1.4% plant dry weight) with growing conditions, particularly climate and geographical location [8]. Furthermore, A. annua flavonoids have been shown to enhance the antiplasmodial activity of artemisinin in vitro [9a-9c]. Therefore, these components should be monitored in locally prepared A. annua herbal drugs in order to assess the quality of the drug used to treat malaria. In this regard, we report herein the HPLC-DAD-MS analyses of different extracts of the aerial parts of A. annua, cultivated in malarial-endemic regions in East Africa. Finally, quantification of the active constituents in these plant materials offered an opportunity to assess the possible role of environmental conditions, such as altitude, in their biosynthesis, as the same A. annua cultivar, Artemis [8], was planted in three different locations in Burundi with distinct geographical and climatic conditions. The applied HPLC-DAD-MS method [9d] was specific for the detection of artemisinin and A. annua flavonoids, even if present only in trace amounts. The plant material used was the cultivar Artemis [8], grown in different locations in Burundi, East Africa, namely Kyenzi, a prairie at 2300 m altitude in central Burundi, J1, a wooded area at ca. 1800 m near the border with Ruanda, and in a field near the hospital of Bubanza, a city situated in north-west Burundi, at 950 m altitude. The n-hexane and dichloromethane extracts of each herbal drug sample were prepared, because it was known from previous investigations [9d] that the extraction efficiency for artemisinin and flavonoids is maximised using these solvents. Samples were analysed by HPLC-DAD-MS. In the extracts, all polymethoxyflavonoids related to the antimalarial activity, such artemetin, chrysoplenetin, casticin, cirsilineol and eupatin were detected. However, as shown in Table 1, yields of each of these constituents varied significantly among Lapenna et al. the different samples. In the n-hexane extracts artemisinin yields ranged from 10.7% to 5.7% (w/w), while in the dichloromethane extracts the total flavonoid content ranged from 12.8% to 5.2%. Table 2 shows the flavonoid variability in the herbal drugs from different cultivations. Finally, table 3 reports the artemisinin and total flavonoid levels expressed as percentage, w/w, of herbal drug. We found that the A. annua plant materials obtained from the different cultivation areas of Burundi contained artemisinin the range 0.20%-0.35% of the herbal drug. These values are inferior to those reported for the same original cultivar (Artemis) after professional cultivation (0.5%-0.75%) [9d]. The lower yields of artemesinin found in the analysed herbal drugs could be a consequence of altered agricultural and collection practices operated at the local sites of production with respect to the established methods for attainment of high-yielding plants [10]. Furthermore, the diverse geographical and climatic conditions, or soil composition of the different fields in Burundi could be responsible for the observed yield variation in A. annua antimalarial constituents. In particular, we noticed that plants grown at higher altitude (i.e. at 2300 m in the Kyenzi region) were richer in artemisinin (0.35%, w/w, of herbal drug) than plants produced at either 1800 m (0.24%) or 950 m (0.20%), in the J1 area and in Bubanza, respectively. The beneficial influence of altitude on artemisinin yields in A. annua plants cultivated at tropical latitudes had been suggested previously [10] and the data collected in this work seems to indicate an analogous relationship. The total flavonoid percentage in the analysed samples ranged from 0.80% to 0.32%, w/w, of herbal drug, with the highest amount present in the samples produced in the fields at higher altitude. In conclusion, we analysed samples of A. annua herbal drug obtained from cultivation at three different sites in Burundi, East Africa. All three samples analysed possessed detectable levels of artemisinin and flavonoids. However, the quantitative profiles of the antimalarial active compounds varied significantly in the different samples. Our results suggest that there could be a correlation between the content of artemisinin and flavonoids and the altitude of the growing site in the East African territory. Further investigations will need to be undertaken in order to assess the best conditions for growing high artemisinin and flavonoid-yielding A. annua plants in these conditions. Comparative analysis of antimalarial principles in Artemisia annua Natural Product Communications Vol. 3 (12) 2008 2005 Table 1: Extraction yield and percentage of artemisinin and flavonoids in the Artemisia annua extracts (dm: dichloromethane, hx: n-hexane) from the Burundi cultivations of J1, Bubanza (Bb) and Kyenzi (Ky). Yield (mg)a 1 2 3 4 5 6 a J1_dm J1_hx Bb_dm Bb_hx Ky_dm Ky_hx 622.0 287.6 762.0 351.8 627.0 329.2 % total flavonoidsb average value 5.2 2.4 6.1 2.1 12.8 3.3 stand. dev. 0.025 0.040 0.026 0.026 0.042 0.042 % artemisininb average value 3.9 8.3 2.6 5.7 4.8 10.7 stand. dev. 0.396 1.048 0.355 0.807 0.632 1.261 Dried extract obtained from 10,0 g herbal drug; bPercentage content in the dried extract. Table 2: Percentage content (w/w) of individual flavonoids in the Artemisia annua herbal drugs from the Burundi cultivations of J1, Bubanza (Bb) and Kyenzi (Ky). Percentage content in the dried extract (w/w); n=3 samples. Extract 1 2 3 4 5 6 J1_dm J1_hx Bb_dm Bb_hx Ky_dm Ky_hx % Eupatin average value stand. dev. 2.49 0.010 0.25 0.003 4.35 0.033 0.19 0.003 2.13 0.027 0.23 0.002 % Cirsilineol average value stand. dev. 0.06 0.003 0.20 0.003 0.10 0.004 - Table 3: Percentage content (w/w) of artemisinin and flavonoids in the Artemisia annua herbal drugs from the Burundi cultivations at J1, Bubanza (Bb) and Kyenzi (Ky). J1 Bb Ky % total flavonoidsa average value stand. dev. 0.32 0.000 0.46 0.000 0.80 0.000 % artemisininb average value stand. dev. 0.24 0.029 0.20 0.023 0.35 0.044 a Percentage of the dichloromethane extract; n=3 samples. bPercentage of the n-hexane extract; n=3 samples. Experimental Chemicals: Artemisinin (98%) was purchased from Sigma (Sigma-Aldrich S.r.l., Milan, Italy) and rutin (99.5%) from Merck (Darmstadt, Germany). Solvents for extraction and HPLC analysis (n-hexane, dichloromethane, MeOH and acetonitrile) were HPLC grade and were purchased from Merck. 85%. Formic acid was purchased from Carlo Erba (Milan, Italy). Water for HPLC analysis was purified by a Milli-Qplus system from Millipore (Milford, MA). Plant material: The seeds of Artemisia annua cv. Artemis were provided by Anamed in Germany (seeds “ANAMED A3”). Cuttings of germinated plants were planted in June 2006 by local farmers at three different locations in Burundi, namely Kyenzi, J1 and Bubanza, in areas exposed to sunlight. Leaves were harvested in November 2006 and dried in the air at temperatures below 40°C. Samples of dry aerial parts were sent to Europe in packages of 100 g. Plant material was cultivated and collected under the supervision of Paolo Monti, working for the Artemisia Project of the Medical Foundation for Africa and supported by the Department for the fight against malaria of the Ministry of Health of Burundi. % Casticin and Chrysoplenetin average value stand. dev. 2.52 0.018 1.95 0.032 7.60 0.050 2.63 0.036 3.55 0.041 1.58 0.055 % Artemetin average value stand. dev. 0.13 0.006 0.25 0.003 0.62 0.016 0.47 0.007 0.28 0.011 0.27 0.010 Preparation of extracts: Artemisia annua dried aerial parts were cut into small pieces and the leaves were separated from the branches and stems. Only leaves and flowering tops (herbal drug) were used for the analyses (Figure 1). Samples of 10 g herbal drug were exhaustively extracted at room temperature by maceration with 100 mL of either n-hexane or dichloromethane for 72 h. The eluates were subsequently dried under vacuum to obtain the crude extracts. HPLC sample preparation: Five mg or each n-hexane or dichloromethane dried extracts were accurately weighed and suspended in acetonitrile (1.0 mL) in a volumetric flask. The suspensions were sonicated for 20 min, then filtered through a cartridge-type filtration unit with a polytetrafluoroethylene (PTFE) membrane (d = 13 mm, porosity 0.45 µm, Lida manufacturing Corp., Kenosha, WI) and immediately injected. HPLC analyses: Artemisinin and flavonoid (Figure 2) contents of the dried extracts were determined by high performance liquid chromatography (HPLC) coupled with mass spectrometer (MS), according to [9d]. HPLC analyses were performed using a HP 1100 Liquid Chromatograph (Agilent Technologies, Palo Alto, CA, USA) equipped with a HP 1040 Diode Array Detector (DAD), an automatic injector, an auto sampler and a column oven, and managed by a HP 9000 workstation (Agilent Technologies, Palo Alto, CA, USA). The HPLC system was interfaced with a HP 1100 MSD API-electrospray (Agilent Technologies). 2006 Natural Product Communications Vol. 3 (12) 2008 Separations were performed on a reversed-phase column of Purospher® Star RP-18, namely Hibar® pre-packed column RT (250 x 4.6 mm), with particle size 5 µm (Merck, Darmstadt, Germany). Eluents were: water adjusted to pH 3.2 with formic acid (A) and acetonitrile (B). The mobile phase was isocratic 50% A and 50% B for 15 min, following by gradient from 50% to 100% B in 5 min, at a flow-rate of 1.0 mL/min. The system was operated with an oven temperature at 26oC; the injection volume was 20 μL. Chromatograms were recorded both at 350 nm to detect the flavonoids and at 210 nm to detect artemisinin and any other constituents, with a peak threshold of 0.1 mAu. The following mass spectrometry operating conditions were used: gas temperature 350°C at a flow-rate of 10 L/min, nebulizer pressure 30 psi, quadrupole temperature 30°C, and capillary voltage 3500 V. Full scan spectra from m/z 100 to 800 in the positive ion mode were recorded (scan time 1 s). Calibration and quantitative analyses: Calibration curves of artemisinin and rutin were obtained from Lapenna et al. stock solutions of each standard in acetonitrile (artemisinin 0.170 mg/mL and rutin 0.092 mg/mL) and used to quantify the artemisinin and flavonoid contents, respectively, in the samples of A. annua extracts. For artemisinin calibration, HPLC injection volumes of 10, 15, 20, 25 and 30 μL of the artemisinin stock solution were used and the peak areas in the MS were recorded. For flavonoid calibration, HPLC injection volumes of 2, 4, 6, and 8 μL of the rutin stock solution were used and the peak areas in the UV chromatogram at 350 nm were measured. Linear regression was used to establish the calibration curve. Each HPLC sample of A. annua extract was injected three times and the artemisinin and flavonoid contents were calculated on the basis of the peak areas in the mass spectra (for artemisinin) or in the UV spectra at 350 nm (for the flavonoids). Acknowledgments - We are thankful to Mr Paolo Monti, responsible for the Artemisia Project in Burundi, for sending us the plant materials. The financial support of Ente Cassa di Risparmio di Firenze is gratefully acknowledged. References [1] (a) Trigg PI, Kondrachine AV. (1998) Commentary: malaria control in the 1990s. Bulletin of the World Health Organization, 76, 11-16; (b) World Health Organization. (2000) Severe falciparum malaria. Transactions of the Royal Society of Tropical Medicine and Hygiene, 94, 1-90; (c) Winstanley P. (2000) Chemotherapy for P. falciparum malaria: the armoury, the problems and the prospects. Parasitology Today, 16, 146-153; (d) Guthmann J-P, Bonnet M, Ahoua L, Dantoine F, Balkan S, Van Herp M, Tamrat A, Legros D, Brown V, Checchi F. (2007) Death rates from malaria epidemics, Burundi and Ethiopia. Emerging Infectious Diseases, 13, 140-143; (e) Checchi F, Cox J, Balkan S, Tamrat A, Priotto G, Alberti KP, Guthmann J-P. (2006) Malaria epidemics and interventions, Kenya, Burundi, southern Sudan, and Ethiopia, 1999-2004. Emerging Infectious Diseases, 12, 1477-1485; (f) World Health Organization. (2001) Weekly Epidemiological Report, 5th January 2001. [2] Van Agtmael MA, Eggelte TA, Van Boxtel CJ. (1999) Artemisinin drugs in the treatment of malaria: from medicinal herb to registered medication. Trends in Pharmacological Sciences, 20, 199–204. [3] Hien T, White N. (1993) Qinghaosu. Lancet, 341, 603-608. [4] Klayman DL. (1985) Qinghaosu (artemisinin): an antimalarial drug from China. Science, 228, 1049-1055. [5] Mueller MS, Karhagomba IB, Hirt HM, Wemakor E. (2000) The potential of Artemisia annua L. as a locally produced remedy for malaria in the tropics: agricultural, chemical and clinical aspects. Journal of Ethnopharmacology, 73, 487–494. [6] Mueller MS, Runyambo N, Wagner I, Borrmann S, Dietz K, Heide L. (2004) Randomized controlled trial of a traditional preparation of Artemisia annua L. in the treatment of malaria. Transactions of the Royal Society of Tropical Medicine and Hygiene, 98, 318-321. [7] Räth K, Taxis K, Walz G, Gleiter CH, Li S-M, Heide L. (2004) Pharmacokinetic study of artemisinin after oral intake of a traditional preparation of Artemisia annua L. American Journal of Tropical Medicine and Hygiene, 70, 128-132. [8] Delabays N. (1997) Biologie de la reproduction chez l’Artemisia annua L. et génétique de la production en artémisinin. Contribution à la domestication et à l'amélioration génétique de l'espèce. Université de Lausanne. [9] (a) Bilia AR, Lazari D, Messori L, Taglioli V, Temperini C, Vincieri FF. (2002) Simple and rapid physico-chemical methods to examine action of antimalarial drugs with hemin. Its application to Artemisia annua constituents. Life Sciences, 70, 769-778; (b) Elford B, Roberts M, Phillipson J, Wilson R. (1987) Potentiation of the antimalarial activity of Qinghaosu by methoxylated flavones. Transactions of the Royal Society of Tropical Medicine and Hygiene, 81, 434-436; (c) Liu KCS, Yang SL, Roberts MF, Elford BC, Phillipson JD. (1989) The contribution of flavonoids to the antimalarial activity of Artemisia annua. Planta Medica, 55, 654-655; (d) Bilia AR, De Malgalhaes PM, Bergonzi MC, Vincieri FF. (2006) Simultaneous analysis of artemisinin and flavonoids of several extracts of Artemisia annua L. obtained from a commercial sample and a selected cultivar. Phytomedicine, 13, 487-493. [10] Ferreira JFS, Laughlin JC, Delabays N, De Magalhães P.M. (2005) Cultivation and genetics of Artemisia annua L. for increased production of the antimalarial artemisinin. Plant Genetic Resources-Characterization and Utilization, 3, 206-229. NPC Natural Product Communications In vitro Apoptotic Bioactivity of Flavonoids from Astragalus verrucosus Moris 2008 Vol. 3 No. 12 2007 - 2012 Joseph A. Buhagiara, Alessandra Bertolib*, Marie Therese Camilleri-Podestac and Luisa Pistellib a Department of Biology, Faculty of Science, University of Malta, MSIDA MSD 06, Malta b Dipartimento di Chimica Bioorganica e Biofarmacia (DCBB), University of Pisa, Via Bonanno 33, Pisa I-56126, Italy c Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, MSIDA MSD 06, Malta bertoli@farm.unipi.it Received: July 30th, 2008; Accepted: November 17th, 2008 Six aglycone flavonoids and their corresponding glycosides: genistein and genistin, quercetin and rutin, apigenin and apigenin 7-O-β-D-(6-p-coumaroyl) glucoside, as well as the aglycone daidzein isolated from Astragalus verrucosus Moris, were tested for their apoptosis-inducing potential. In vitro techniques that monitor bioactivity through morphological and biochemical changes were carried out on HCT116 (human colon carcinoma) and MCF7 (human Caucasian breast adenocarcinoma) cancer cell lines. Dose-dependent cytotoxic effects were monitored through changes in mitochondrial dehydrogenase activity using the standard MTT assay. The median inhibitory concentration (GI50) determined from the dose-response curves showed that the aglycones apigenin and quercetin were the most bioactive (low GI50), whilst daidzein and genistein, which had not been previously tested on these cell lines, showed a smaller cytotoxic effect (high GI50). The remaining flavonoids, mostly glycosides, showed negligible cytotoxicity. Morphological changes were monitored by microscopic observation with a photographic record. Results showed important hallmarks of apoptosis, including cell rounding with reduction of cell volume, small condensed nuclei, membrane blebbing and formation of apoptotic bodies. Keywords: Flavonoids, aglycones, glycosides, apoptosis, cancer cell lines, HCT116, MCF7. Astragalus verrucosus Moris, a very rare perennial endemic plant belonging to the family Fabaceae (Leguminosae), grows in a restricted area of southwestern Sardinia, Italy [1]. The genus Astragalus is well known in Chinese folk medicine because its properties are somewhat similar to those of the more expensive herb ginseng (Panax ginseng) and have been used as a substitute for this species [2]. The roots of various Astragalus species have been used to increase body resistance against viral infections, to re-balance the immune system and for their tonic action on the liver. Extracts from Astragalus species have been used as antiperspirant, diuretic and general tonic agents. The roots have been applied in the treatment of diabetes mellitus, nephritis, and bacterial infection, as well as against leukaemia and uterine cancer [3,4]. Phytochemical studies of A. verrucosus have shown the presence of several classes of flavonoids, such as flavonols, isoflavones, and flavones, both as aglycones and glycosides (Table 1). Over 6000 different flavonoids (aglycones) have been identified to date, and this figure increases considerably if their corresponding glycosides are included [6-8]. Flavonoids are plant secondary metabolites, especially widespread in the plant kingdom and ubiquitous in photosynthesising cells. Flavonoids represent important dietary constituents because of their antioxidant activity. They are accumulated in different plant parts including fruits, vegetables, nuts, seeds, and flowers, as well as in products derived from them, such as propolis, honey and tea [5]. Flavonoids are well known for their different pharmacological properties, including: anti-allergic, immunoregulatory, antioxidant, anti-inflammatory, hypotensive, antibacterial, antifungal, antiviral, cytotoxic and osteogenic activities [6-10]. Preparations containing these compounds have been used to treat different diseases for centuries. Some are borrowed directly from nature, as for example propolis, which has been used since antiquity to heal 2008 Natural Product Communications Vol. 3 (12) 2008 Table 1: Typical flavonoids of Astragalus verrucosus FLAVONES apigenin apigenin 7-O-β-D(6”-p-coumaroyl)glucoside FLAVONOLS kaempferol 3-O-rutinoside kaempferol 3-robinobioside rutin quercetin ISOFLAVONES daidzein daidzin ononin calycosin pseudobaptigenin genistein genistin pratensein REF. [19] [19] [19] [24] [15] [4] [19] [19] [25] [17] [15] [19] [19] [19] sores and wounds. Several flavonoid-rich foods (such as quercetin-rich apples) have been reported to lower the risk of different types of cancer, including hepatoma, lung and breast cancer [8]. Flavonoids such as quercetin have been reported to inhibit the growth of various cell lines derived from human cancers [6,9]. They have been shown to be selective cytotoxic agents to a range of cancer cell lines, including Jurkat, PC-3, colon 205 and HepG2. The mode of cell death in the human promyelocytic leukaemia HL-60 cell line treated with the structurally related flavonoids apigenin, quercetin, myricetin, and kaempferol has been established as being apoptotic through the release of cytochrome C, as well as caspase-9 and caspase-3 into the cytosol [7,11]. Other flavonoids have been studied and shown to be capable of inducing cell death by apoptosis [12]. Interestingly, quercetin was found to restore apoptosis sensitivity in cell lines usually resistant to apoptosis [13]. Terminal differentiation has also been identified as another possible mechanism of flavonoid action and, therefore, these compounds can serve as potential chemotherapeutic agents [14]. Most plant-derived natural products are often produced as a mixture of related compounds that have a synergistic mode of action, either among themselves or with other compounds, and flavonoids are no exception. The synergistic effects of three isoflavones: genistein, biochanin-A and daidzein have also been demonstrated in cancer cell lines through cell growth inhibition, cell cycle changes, and induction of apoptosis. Enhanced expression of pro-apoptotic caspase 3 and downregulation of antiapoptotic Bcl-2 was also demonstrated [8,11,14]. Of increasing interest also is the potential role of flavonoids in reducing the problems of drug resistance to antimicrobials and in cancer therapy. This has also been extensively investigated and holds Buhagiar et al. some promise, especially with the increasing problems associated with β-lactamase resistance and MRSA [6,7]. This current work was addressed to test the bioactive potential and apoptosis-inducing activity of some characteristic flavonoids isolated from A. verrucosus on HCT-116 and MCF-7 tumor cell lines. These compounds include two aglycones (daidzein and genistein) that have not been previously tested on these cell lines. Although work on the induction of apoptosis in cancer cells by flavonoids appears to be gaining momentum, there is still insufficient published data on the action of the various flavonoids on different cell types. The NCI database gives the average GI50 values for its 50 cancer cell line panel as 27 μM for apigenin and 59 μM for quercetin. Though the values for the individual cell lines are not available, these average values confirm the trend that we obtained for the same test compounds in this work. This research has also confirmed the trend in the published literature that the aglycone flavonoids are generally more potent than their corresponding glycosides. What seems to be omitted in the literature is a reference to the paradoxical increase in cell proliferation on exposure to low concentrations of the aglycones, something which has also been consistently observed with another group of natural compounds, the monosesqui- and di-terpenoids. Flavonoid molecules have structural similarities and comparable molecular sizes to the diterpenoids. Like diterpenoids, aglycone flavonoid molecules are planar and have been shown capable of interacting, to various degrees, with the phospholipid bilayer. The degree of flavonoid interaction has been shown to vary according to whether hydroxyl groups are present or absent, the number of hydroxyl groups and the position of their attachment to the A and B ring [5,15]. Apart from the chemical interactions of the side groups, the small size of the aglycones allows for a greater chance of interacting deep within the inner layers of the phospholipid bilayer and brings about changes in membrane fluidity. Such changes in membrane fluidity as a result of flavonoid interactions have been reported both for prokaryotic and eukaryotic cells, as a result of which cells lose important capabilities. For instance, flavonoid action on the inner membrane of Gram-positive bacteria has been reported to lead to dissipation of membrane potential electrochemical gradient with consequent reduction of ATP synthesis, membrane transport and motility. In other experiments, the action of flavonoids leads Bioactivity of flavonoids from Astragalus verrucosus Natural Product Communications Vol. 3 (12) 2008 2009 to increased permeability of the inner membrane with loss of important cell constituents such as K+ ions [5]. A similar effect is reported for a number of flavonoids, including quercetin; this has been shown to interact with the membrane of the mitochondria, decreasing its fluidity and, as a result, either inhibiting the respiratory chain or inducing membrane permeability transition [15]. The latter is equivalent to a state where the mitochondria lose their function and release apoptosis-inducing factors that lead to cell death by apoptosis. membranes by natural products such as flavonoids could explain why apoptosis results when cancer cells are treated with these compounds. A change in membrane fluidity (especially of the mitochondrial membrane) could lead to the release of pro-apoptotic signals from the mitochondria, such as cytochrome c, and trigger the apoptotic cascade which eventually results in cell death. Experimental Extraction, flavonoid isolation and identification: Astragalus verrucosus dried aerial parts were extracted in a Soxhlet apparatus using different solvents with increasing polarity (n-hexane, chloroform, and methanol) [4]. The methanolic extract was purified by gel permeation (Sephadex LH-20, MeOH-Water 8:2) and medium pressure liquid chromatographic steps (SiO2 RP-9, MeOH-water 7:3). Seven flavonoids: apigenin, apigenin 7-O-β-D-(6p-coumaroyl) glucoside, quercetin, rutin, daidzein, genistein and genistin were isolated and identified by NMR and MS experiments, and by comparison with authentic samples and literature data [4,18,19]. Table 1 gives additional data on other flavonoids isolated from A. verrucosus [21,22]. The aglycone flavonoids that have been shown to be the most bioactive represent relatively small molecules that can easily interact with the cell membrane. Conversely, since the glycoside flavonoids represent a bulkier molecule, of which only a part can interact with the phospholipid bilayers, they are unable to interact deeply enough inside the membrane to cause drastic changes in fluidity. This hypothesis could neatly explain the differences observed in the action of different aglycone and glycoside flavonoids where for the glycosides, a relatively flat dose-response curve was maintained, even at high concentrations, indicating that interaction does not increase, even under an appreciable concentration gradient. Cell lines and medium: Two adherent cell lines, namely human colon carcinoma cell line HCT 116 and human Caucasian breast adenocarcinoma cell line MCF 7, were obtained from ECACC Porton Down, Salisbury, UK. RPMI-1640 medium with 2 mM L-glutamine and 1mM sodium pyruvate (Gibco BRL, Life Technologies) was supplemented with 10% fetal bovine serum (Gibco) and 25 IU/mL penicillin G and 25 µL/mL streptomycin (PenStrep, Gibco). All cell lines were kept in exponential growth phase by twice weekly subculture in T25 cell culture flasks (Nunc, Kampstrum, Denmark) in 6 mL of medium using a split ratio of 1:5. The relevance of the role of aglycone flavonoids in the induction of apoptosis in cancer cells has to be considered in the light of emerging research as to the role of mitochondria in cancer. Formerly, it was thought that cancer cells are predominantly glycolytic because their mitochondria were defective and incapable of generating the vast quantities of ATP needed to sustain growth of cancer cells. However, the evidence points to fully functional mitochondria that are pushed into an inactive state so as to suppress the apoptotic cascades that are normally initiated in mitochondria [16,17]. Thus, this perturbation of Table 2: Results of apoptotic activity in HCT116 and MCF7 cancer cell lines for the flavonoids isolated from the methanolic extract of Astragalus verrucosus. All median inhibitory concentration values (GI50) shown are an average of at least three replicates. COMPOUNDS Quercitin Daidzein Apigenin Genistein Rutin apigenin -7-O-β-D-(6’’-pcoumaroyl)-glucoside genistin replicate 1 9.0 76.8 5.9 33.2 >100 >100 HCT116 2 20.9 >100 3.3 >100 >100 >100 (GI50) 3 28.8 >100 2.8 53.9 >100 >100 Average (µg/mL) 19.6 76.8 (>100) 4.0 43.5 (>100) >100 >100 >100 >100 >100 >100 replicate 1 >100 66.2 5.7 60.2 >100 >100 MCF7 (GI50) 2 3 47.2 9.8 >100 >100 4.2 4.0 37.8 64.9 >100 >100 >100 >100 >100 >100 >100 Average (µg/mL) 28.5 (>100) 66.2 (>100) 4.6 54.3 >100 >100 >100 2010 Natural Product Communications Vol. 3 (12) 2008 Buhagiar et al. Figure 1: Dose-response curves for 48 h exposure of HCT116 (top) and MCF7 (bottom) cancer cell lines treated with various concentrations of aglycone flavonoids (left) and their corresponding glycosides (right). All values are plotted as a percentage of control absorbance values. Absorbance values were measured at 550 nm using the standard MTT photometric assay. MTT assay: Cell cultures in exponential growth phase were trypsinised with 0.25% trypsin in EDTA, and after a viable cell count diluted to give a seeding density of 5000 cells per 180 µL of culture medium. The cells were then plated in flat-bottomed 96-well micro titer plates (Nunc) and incubated overnight at 37°C in a 5% CO2 humidified atmosphere. A time zero (T0) reference plate was also set up to check cell viability at the point of first drug exposure. The appropriate dilutions of the flavonoids in culture medium were prepared from a stock solution of 40µg/µL in DMSO and 20µL aliquots of the drug were added per well to give a final concentration range from 0.01 μg/mL to 100 μg/mL, plus solvent controls. Plates were further incubated for a maximum of 44 h, with inspection at regular intervals to check growth progress. Cell viability was determined by the addition of a 50 µL aliquot of MTT (2.0 mg/mL) per well, and insoluble formazan crystals allowed to develop by incubating for a further 4 h. Formazan was dissolved by adding 125 mL of solubilization Sorensen’s buffer (DMSO : glycine 4:1) to each well and the plates were agitated for 5 minutes. The absorbances were read using a microplate reader (BioTech ELx 808) at 550 nm and 650 nm wavelength. Dose-response curves were generated using a Delta soft 3 software package and in combination with the T0 value, the median growth inhibition (GI50) was determined for three independent trials. Absorbance values derived from the MTT assays were used to generate dose-response curves, from which the median inhibitory concentration (GI50) was calculated. The average GI50 values (μg/mL) for three independent trials are shown in Table 2. The cell growth was also plotted as a percentage of the control, and typical graphs are shown in Figure 1. Study of morphological changes: These were restricted to a photographic record of the changes that were observed in the overall cell structure when visualised under an inverted microscope at high magnification. Photos were taken after 24 and 44 h exposure before adding the MTT. The doseresponse curves for the aglycone flavonoids were typical of cytotoxic drugs with GI50 values in the range of 4-76 µg/mL. The highest activity of the aglycones was for apigenin. Furthermore, its GI50 values are very similar in the two cell lines tested with an average GI50 for HCT116 of 4.0 µg/mL and Bioactivity of flavonoids from Astragalus verrucosus Natural Product Communications Vol. 3 (12) 2008 2011 Figure 2: HCT 116 and MCF7 cells after 48 h treatment with quercitin (10 and 50 µg/mL) with solvent controls (top). for MCF7 of 4.6 µg/mL. The bioactivity of quercetin was the next best with an average GI50 value in the 20 µg/mL range for HCT116 and 28.5 µg/mL for MCF7. The largest GI50 values for the aglycones and, therefore, the least bioactive were for genistein and daidzein. Conversely, the dose-response curves of their corresponding glycosides were in most cases rather flat and showed large GI50s in excess of 100 µg/mL, indicating that there is little cytotoxic effect on these two cell lines. The poor bioactivity of the glycoside derivatives was also corroborated from the morphological observations. One interesting feature of the dose-response curves of some of the aglycones and glycoside flavonoids was that at very low concentrations, a paradoxical stimulation of growth occurred, sometimes reaching 130% of the control. This is usually followed by a steep decrease in activity as the concentration increases. This paradoxical increase in cell activity for very low concentrations has been reported for other natural products including mono- and diterpenoids and may be related to their as yet unexplained mode of action [23]. Morphological changes resulting from the exposure of cells to different types of flavonoids and different time periods demonstrated that the cells undergo apoptosis with increasing concentration and with increased exposure time. Figure 2 shows the two cell lines treated at the two concentrations (10 and 50 µg/mL) with solvent controls after 48 h exposure, although the full exposure included a concentration range from 1-100 µg/mL. These morphological changes were clearly demonstrated under the high power objective (x400) without the need of further staining since these changes were comparable to those obtained in previous works [23]. The major hallmarks were the loss of cellular extensions and substrate contact resulting in the formation of round, free floating cells, some with extensive membrane blebbing and formation of apoptotic bodies. A reduction in overall cell volume resulting from cytoskeletal disruption and the formation of a small pycnotic nucleus from DNA condensation were also noted. For the active flavonoids, changes such as partial loss of contact with substrate and rounding off were initially observed at 10 µg/mL after 24 hour 2012 Natural Product Communications Vol. 3 (12) 2008 exposure, progressing to pronounced hallmarks of apoptosis, such as membrane blebbing and nuclear condensation as concentration increased. At Buhagiar et al. 50 µg/mL the cells were completely spherical and lost all attachment to the substrate after the same exposure time. References [1] Pignatti S. (1982) Flora d’Italia. Agricole Ed., Italy, pp.652. [2] Tang W, Eisenbrand G. (1992) Chinese drugs of plant origin, Springer-Verlag, Berlin. [3] Pistelli L. (2002) Secondary metabolites of genus Astragalus: structure and biological activity. In Studies in Natural Products Chemistry, Vol. 27, Bioactive Natural Products (Part H) Atta-Ur-Rahman (Ed.), Elsevier, Amsterdam, pp.443-455. [4] Pistelli L, Giachi I, Lepori E, Bertoli A (2003) Further saponins and flavonoids from Astragalus verrucosus Moris,. Pharmaceutical Biology, 41, 568-572. [5] Scheidt AH, Pampel A, Nissler L, Gebhardt R, Huster D. (2004) Investigation of the membrane localization and distribution of flavonoids by high-resolution magic angle spinning NMR spectroscopy. Biochimica et Biophysica Acta, 1663, 97-107. [6] Middleton EJr, Kanaswami C, Theohardarides TC. (2000) The effects of plant flavonoids on mammalian cells: implications for inflammation, heart disease, and cancer. Pharmacological Reviews, The American Society of Pharmacological and Experimental Therapeutics, pp. 673-751. [7] Xu HX, Lee SF (2001) Activity of plant flavonoids against antibiotic-resistant bacteria. Phytotheraphy Research, 15, 39-43. [8] Su SJ, Chow NH, Kung ML, Hung TC, Chang KL (2003) Effects of soy isoflavons on apoptosis induction and G2-M arrest in human hepatoma cells involvment of caspase-3 activation, Bcl-2 and Bcl-XL downregulation, and Cdc2 kinase activity. Nutrition and Cancer, 45, 113-123. [9] Avila M, Cansado J, Harter K, Velasco J, Notario V. (1996) Quercetin as a modulator of the cellular neoplastic phenotype, Advances in Experimental Medicine and Biology, 401, 401-410. [10] Kyle J, Duthie, G. (2006) Nutritional relevance of flavonoids in disease prevention. Natural Product Communications, 1, 1049-1060. [11] Wang IK, Lin-Shiau SY, Lin JK. (1999) Induction of apoptosis by apigenin and related flavonoids through cytochrome c release and activation of caspase-9 and caspase-3 in leukaemia HL-60 cells. European Journal of Cancer, 35, 1517-1525. [12] Russo M, Palumbo R, Tedesco I, Mazzarella G, Russo P, Iacomino G, Russo GL (1999) Quercetin and anti-D95(Fas/Apol) enhance apoptosis in HPB-ALL cell line, FEBS Letters 462, 322-328. [13] Russo M, Palumbo R, Mupo A, Tosto M, Scognamiglio A, Tedesco I, Galano G, Russo P, Iacomino G, Russo GL (2003) Flavonoid quercetin sensitizes a CD95-resistant cell line to apoptosis by activating protein kinase Cα, Oncogene, 22, 3330-3342. [14] Ren W, Qiao Z, Wang H, Zhu L, Zhang L. (2003) Flavonoids: promising anticancer agents, Medicinal Research Reviews, 23, 519-534. [15] Dorta DJ, Pigoso AA, Mingatto FE, Rodrigues T, Prado IMR, Helena AFC, Uyemura SA, Santos AC, Curti C. (2005) The interaction of flavonoids with mitochondria: effects on energetic processes, Chemico-Biological Interactions, 152, 67-78. [16] Bensaad K, Tsuruta A, Selak MA, Vidal MNC, Nakano K, Bartrons R, Gottlieb E, Vousden K. (2006) TIGAR, a p53-inducible regulator of glycolysis and apoptosis. Cell, 126, 107-120. [17] Garber K. (2006) Energy dysregulation: licensing tumours to grow. Science, 312, 1158-1159. [18] Markham KR, Mabry TJ (1968) The identification of twenty-three 5-deoxy and 5-hydroxy-flavonoids from Baptisia lecontei (Leguminosae). Phytochemistry, 7, 791-801. [19] Harborne JB, Baxter H (1999) The Handbook of Natural Flavonoids, Vol I - II, J. Wiley & Sons, Chichester, [20] Agrawal PK, Markham KR (1989) Carbon-13 NMR of Flavonoids, Agrawal PK (Ed), Elsevier Science Publishers B.V., The Netherlands, pp. 194-208. [21] Rao LJM, Kumari GNN, Rao NSP (1985) Flavonoid glycosides from Anisomeles ovata. Journal of Natural Products, 48, 150-151. [22] Cui B, Sakai Y, Takeshita T, Kinjo J, Nohara T. (1992) Four new oleanene dervatives from the seeds of Astragalus complanatus. Chemical & Pharmaceutical Bulletin, 40, 136-138. [23] Buhagiar JA, Camilleri Podesta MT, Wilson AP, Micallef MJ, Ali S. (2000) The induction of apoptosis in human melanoma, breast and ovarian cancer cell lines using an essential oil extract from the conifer Tetraclinis articulata. Anticancer Research, 19, 5435-5444. [24] Nawwar MAM, El-Mousallamy AMD, Barakat HH, Buddrush J, Linscheia M. (1989) Flavonoid lactates from leaves of Marrubium vulgare. Phytochemistry, 28, 3201-3206. [25] Rao LJM, Kumari GNN, Rao NSP (1985) Flavonoid glycosides from Anisomeles ovata. Journal of Natural Products, 48, 150-151. NPC Natural Product Communications Qualitative Profile and Quantitative Determination of Flavonoids from Crocus sativus L. Petals by LC-MS/MS 2008 Vol. 3 No. 12 2013 - 2016 Paola Montoroa, Carlo I. G. Tuberosob, Mariateresa Maldinia, Paolo Cabrasb and Cosimo Pizzaa a Dipartimento di Scienze Farmaceutiche, Università di Salerno, Via Ponte Don Melillo, 84084 Fisciano (SA), Italy b Dipartimento di Tossicologia, Università di Cagliari, via Ospedale 72, 09124 Cagliari, Italy pmontoro@unisa.it Received: June 10th, 2008; Accepted: October 16th, 2008 From the methanolic extract of Crocus sativus petals nine known flavonoids have been isolated and identified, including glycosidic derivatives of quercetin and kaempferol as major compounds (1-2), and their methoxylated and acetylated derivatives. Additionally, LC-ESI-MS qualitative and LC-ESI-MS/MS quantitative studies of the major compounds of the methanolic extract were performed. The high content of glycosylated flavonoids could give value to C. sativus petals, which are a waste product in the production of the spice saffron. Keywords: Crocus sativus, LC-ESI-MS, LC-MS/MS, quercetin, kaempferol. Saffron, the dried stigmas of Crocus sativus L. (Iridaceae), is a very expensive spice, and is used as a herbal medicine, for food coloring and as a flavoring agent in different parts of the world [1a]. Saffron originally grew in India, Iran, Europe and other countries, and it has been successfully cultivated in different countries, including Europe. The most important European production areas are Sardinia and Abbruzzo (Italy), Castile-la Mancha (Spain) and western Macedonia (Greece). For saffron, the flowers are cultivated to produce the stigmas. After harvesting, the flowers are subjected to a delicate treatment which will give the saffron spice. This procedure is performed the same day of harvest. One of the most traditional procedures is the separation of petals from stigmas. A large amount of petals is discarded for obtaining a small amount of stigmas. Earlier investigation reported the isolation of carotenoids, crocins, monoterpenoids and flavonoids from the stigma, leaves, petals and pollen of C. sativus [1b,1c]. Considering the large amount of petals that are waste products in this production procedure, we have undertaken a study to recover chemical compounds from this matrix. Only one previous paper concerning petals is reported in the literature, oriented towards the biological activity of some new phenols isolated from this part of the flowers [2a]. Flavonoids are polyphenolic compounds with antioxidant properties [2b,2c]. Several studies have shown that a high intake of flavonoids has been correlated to a decrease in heart disease; in addition, biological effects of this class of compounds have been described in several in vivo and in vitro studies [3a-3d]. These compounds are largely used for chemotaxonomic surveys of plant genera and families because of their almost ubiquitous presence in vascular plants and of their structural variety. A phytochemical study was undertaken with the aim of identifying and determining quantitatively the major compounds in the petals. In this study flavonoid compounds were isolated; the major compounds were glycosidic derivatives of quercetin and kaempferol, including their methoxylated and acetylated derivatives (1-9). The study provided a method to define the flavonoids fingerprint by LC-ESI- IT MS/MS (liquid chromatography electrospray tandem mass spectrometry with ion trap analyser), with full scan acquisition in data dependent scan mode, and a method to quantify the content of the major compounds by LC-ESI- TQ 2014 Natural Product Communications Vol. 3 (12) 2008 MS/MS (liquid chromatography electrospray tandem mass spectrometry with triple quadrupole analyser) by using a MRM (multiple reaction monitoring) mode. The quantitative method, performed by using internal and external standards, was validated in agreement with EMEA note guidance on validation of analytical methods [4]. By using tandem in time mass spectrometry it was possible to reveal the compounds on the basis of their specific fragmentation. The specific fragmentation pattern was used for developing the selective MS/MS method for the two major compounds (1, 2). The use of tandem mass spectrometry in quantification of secondary metabolites from plants has led to sensitive, selective and robust methods for quality control and standardisation of plant extracts [5a-5c]. Phytochemical investigation of the methanolic extract of C. sativus led to the isolation of flavonoid compounds 1-9. The compounds, identified by comparing their NMR data with those reported in the literature, were quercetin-3,7-di-O-β-Dglucopyranoside (1) [6], kaempferol-3,7-di-O-β-Dglucopyranoside (2) [2a], isorhamnetin-3,7-di-O-β-Dglucopyranoside (3) [7], kaempferol-3-O-β-Dglucopyranoside (4) [2a], quercetin-3-O-β-Dglucopyranoside (5) [8], isorhamnetin-3-O-β-Dglucopyranoside (6) [9], kaempferol-7-O-β-Dglucopyranoside (7) [2a], kaempferol-3-O-β-D-(2-Oβ-D-glucosyl) glucopyranoside (8) [2a], and kaempferol-3-O-β-D-(2-O-β-D-6-O-acetylglucosyl) glucopyranoside (9) [2a]. The high content of glycosylated flavonoids could give value to C. sativus petals, which are a waste product in the production of saffron spice. In order to realise a qualitative analysis for the flavonoid derivatives in C. sativus extracts, MS experiments were performed by using an LC-MS system equipped with an ESI source and an Ion Trap analyser. Positive ion electrospray LC/MS analysis, total ion current (TIC) profile and reconstructed ion chromatograms (RICs) of extract are shown in Figure 1. Flavonoid derivatives were identified by comparing retention times and m/z values in the total ion current chromatogram with those of the selected standards, obtained in the isolation step. Reconstructed ion chromatograms were obtained for each value of m/z observed for the standard compounds (m/z 627, 1; m/z 611, 2; m/z 641, 3; m/z 449, 4, 7; m/z 465, 5; m/z 479, 6; and m/z 653, 9) in order to improve the separation and identification of Montoro et al. single compounds. The chromatographic profile obtained in Total Ion Current revealed two very major compounds, respectively 1 and 2, in C. sativus extracts, whereas the other compounds were present in lesser amounts. Quantitative analysis was focused on compounds 1 and 2, which could potentially be recovered from discarded petals as economic secondary products. In order to obtain an accurate quantitative determination of compounds 1-2, a quantitative LCMS/MS method was developed. Since the sugar loss is the most representative fragmentation for glycosidic flavonoids, ESI-MS/MS analyses were recorded for the two major compounds by using an LC-MS equipped with an ESI source and a triple quadrupole analyser. Analyses were performed by direct introduction and both the spectra showed the characteristic fragment resulting from sugar loss. Thus an MRM method was developed. Transition from the specific pseudomolecular ion [M+H]+ of each compound to the corresponding aglycon ion [A+H]+ was selected to monitor the flavone glycosides in C. sativus using as internal standard (I.S.), rutin (m/z 611) (I.S.). Compound 1: precursor ion m/z 627.0, product ion m/z 303.0, collision energy 30%; compound 2: precursor ion m/z 611.0, product ion m/z 287.0, collision energy 30%; I.S. precursor ion m/z 611.0, product ion m/z 303.0, collision energy 30%. The MRM analyses of C. sativus methanolic extract, spiked with I.S., contained the peaks corresponding to the compounds under investigation, with appreciable intensity for quantitative purposes. Validation of the method was realised in agreement with EMEA note guidance on validation of analytical methods [4]. Validation of the LC/MS/MS method included intra and inter-day precision and accuracy studies on three days. Accuracy and precisions were calculated by analysing five samples of each extract (MeOH and water). Standard deviations calculated in this assay were < 7% for the two compounds under investigation. Specificity is usually reported as the non interference with other substances detected in the region of interest; the present method, developed by using a characteristic fragmentation of flavone glycosides 1, 2, was specific with no other peak interfering in the MS/MS detection mode. Flavonoids from Crocus sativus petals 1.13 10 5 26.45 3.22 0 8.65 12.8 21.9 17.18 25.55 36.09 37.8 Base Peak ESI Full ms [ 200.00-1000.00] Equipment: Semi-preparative HPLC was performed using an Agilent 1100 series chromatograph, equipped with a G-1312 binary pump, a G-1328A rheodyne injector and a G-1365B multiple wave detector. The column was an RP C18 column μ-bondapak 300 mm x 7.6 mm (Waters, Milford, MA). HPLC–ESI-MS analysis was performed using a Thermo Finnigan Spectra System HPLC coupled with an LCQ Deca ion trap. Chromatography was performed on an RP C18 column Symmetry Shield (Waters, Milford, MA). HPLC–ES-MS/MS for quantitative analysis was performed on a 1100 HPLC system (Agilent, Palo Alto, CA) coupled with a triple quadrupole instrument [API2000 (Applied Biosystems, Foster City, CA, USA)]. The instrument was used in the tandem MS mode, with multiple reaction monitoring (MRM). 1.13 10 1 21.9 5 20.32 0 1.13 10 m/z= 626.50-627.50 26.45 5 0 1.16 10 21.1 5 m/z= 640.50-641.50 26.4 1.13 10 m/z= 610.50-611.50 2 3 0 7 5 4 m/z= 448.50-449.50 19.5 0 1.13 10 5 5 0 21.9 m/z= 464.50-465.50 20.29 0.99 10 21.1 5 10 30.73 Natural Product Communications Vol. 3 (12) 2008 2015 m/z= 478.50-479.50 6 1.16 0 m/z= 652.50-653.50 9 26.3 5 0 0 5 10 15 20 Time (min) 25 30 35 Figure 1. HPLC-MS qualitative analysis Table 1: Quantitative results and quantification data. n 1 2 mg/g MeOH ext. 41.8±6.2 31.1±4.7 mg/g water ext. 8.1±0.2 5.5±0.1 mg/g petals 27.6±6.0 20.2±4.6 Calibration equation y = 0.183x-0.74 y = 0.094x-0.39 r2 0.998 0.997 The calibration graphs, obtained by plotting area ratio between external and internal standards versus the known concentration of each compound, were linear in the range of 1-100 μg mL-1 for all compounds. Correlation values (r2) are reported in Table 1. Five aliquots of methanol and water extracts, respectively, obtained from C. sativus were analysed in order to quantify the flavonoid contents. Table 1 reports the quantitative data for compounds 1-2, regression of calibration curves, and quantitative values. Quantitative analyses results confirmed that waste petals of C. sativus can represent an interesting source of such phenolic compounds, with respect to the high content showed. Experimental Reagent and standards: Standards of pure compounds 1-2 were isolated in our laboratory and their structures were elucidated by NMR spectroscopy (Bruker DRX-600). Each standard was dissolved in methanol. HPLC grade methanol (MeOH), acetonitrile (ACN) and trifluoroacetic acid (TFA) were purchased from Merck (Merck KGaA, Darmstadt, Germany). HPLC grade water (18mΩ) was prepared using a Millipore Milli-Q purification system (Millipore Corp., Bedford, MA). The reagents used for the extractions, of analytical grade, were purchased from Carlo Erba (Rodano, Italy). Column chromatography was performed over Sephadex LH20 (Pharmacia, Uppsala, Sweden). LC-ESI-MS and LC-ESI-MS/MS analysis: The mass spectrometer was operated in the positive ion mode under the following conditions: capillary voltage 3 V, spray voltage 5 kV, tube lens offset 40 V, capillary temperature 260°C, and sheath gas (nitrogen) flow rate 60 arbitrary units. Data were acquired in the MS1 scanning mode with scan ranges of 200 – 1000 m/z: the maximum injection time was 50 ms, and the number of microscans was 3. In order to tune the LCQ for flavonoids, the voltages on the lenses were optimised using the TunePlus function of the Xcalibur software in the positive ion mode whilst infusing a standard solution of quercetin (1 μg mL-1 in methanol) at a flow rate of 3 μL min-1. For qualitative LC-ESI-MS analysis, a gradient elution was performed on a RP C18 column Symmetry shield (Waters, Milford, MA), 2mm x 150 mm, by using a mobile phase A represented by water acidified with trifluoroacetic acid (0.05%) and a mobile phase B represented by water: acetonitrile 50:50 acidified with trifluoroacetic acid (0.05%). The gradient started from 20% of eluent B, to achieve the 33% of solvent B in 18 min. After another 12 min the percentage of B became 40%, and remained at this value for 10 min, then became 50%. The flow (250 μL min-1) generated by chromatographic separation was directly injected into the electrospray ion source. MS were acquired and elaborated using the software provided by the manufacturer. For quantitative LC-ESI-MS/MS a gradient elution was performed by using a mobile phase A represented by water acidified with trifluoroacetic acid (0.05%) and a mobile phase B represented by acetonitrile acidified with trifluoroacetic acid 2016 Natural Product Communications Vol. 3 (12) 2008 Montoro et al. Plant material: Petals of C. sativus, discarded by production companies of saffron spice, were collected in Sardinia (Italy) in November 2004. lyophilized to give 3 g of crude extract. Part of the methanolic extract (3.7 g) was fractionated initially on a 100 cm×5.0 cm Sephadex LH-20 column, using CH3OH as mobile phase, and 56 fractions (10 mL each) were obtained. Fractions 28-29 (24.5 mg) (a), 19-20 (250 mg) (b), 31-33 (31.4 mg) (c) and 36-40 (40.4 mg) (d) were chromatographed by HPLC-UV. The mobile phase was a linear gradient of water/acetonitrile (50:50) with trifluoroacetic acid 0.1% (solvent B) in water acidified with trifluoroacetic acid 0.1% (solvent A), at a flow rate of 2.000 mL min-1. From sample a, compounds 1 (3.3 mg, tR=26.8), 2 (2.9 mg, tR=29.7) and 3 (1 mg, tR=28.7) were obtained; from sample c, compounds 4 (1.6 mg, tR=36.4) and 6 (0.9 mg, tR=37.8); and from sample d, compounds 5 (0.6 mg, tR=32) and 7 (1.1 mg, tR=36.9) using the following gradient: 0 min, 10% B, 0-5 min, 10-20% B, 5-25 min, 20-40% B; 25-40 min, 40% B; 40-50 min, 40-70% B, 50-60 min, 70-100% B. Extraction and isolation: Dried and powdered petals (23 g) of Crocus sativus were extracted for 3 days, 3 times, at room temperature with methanol to give 9.7 g of crude methanolic extract. This was extracted for one day with water. The filtered extract was From sample b, compounds 8 (5.6 mg, tR=29.8) and 9 (1.2 mg, tR=37.9) were obtained using the following gradient: 0 min, 20% B; 0-18 min, 20-33% B; 18-30 min, 33-40% B; 30-40 min, 40% B; 40-45 min, 40-85% B; and 45-55 min, 85-100%. (0.05%). The gradient started from 5% of eluent B, remained isocratic for 5 minutes, to achieve the 80% of solvent B in 15 min. The flow (250 μL min-1) generated by chromatographic separation was directly injected into the electrospray ion source. The mass spectrometer was operated in the positive ion mode under the following conditions: declustering potential 200 eV, focusing potential 155 eV, entrance potential 10 eV, collision energy 30 eV, and collision cell exit potential 15 eV, ion spray voltage 5000, temperature 250°C. The instrument was used in the tandem MS mode with multiple reaction monitoring (MRM). For all flavonoids analyzed, the selected fragmentation reaction was the loss of the glycoside moiety. References [1] [2] [3] [4] [5] [6] [7] [8] [9] (a) Fernandez, JA. (2004) Biology, biotechnology and biomedicine of saffron. Recent Research Developments in Plant Science, 2 127-159; (b) Rios JL, Recio MC, Giner RM, Manets S. (1996) An update review of saffron and its active constituents. Phytotheapy Research, 10, 189-193; (c) Xi L, Qian Z. (2006) Pharmacological properties of crocetin and crocin (digentiobiosyl esters of crocetin) from saffron, Natural Product Communications, 1, 65-75. (a) Li CY, Lee EJ, Wu TS. (2004) Antityrosinase principles and constituents of the petals of Crocus sativus. Journal of Natural Products, 67, 437-440; (b) Rice-Evans CA, Miller NJ, Paganga G. (1997) Antioxidant properties of phenolic compounds. Trends in Plant Science, 2, 152-159. (a) Cao G, Sofic E, Prior RL. (1997) Antioxidant and pro-oxidant behavior of flavonoids: structure-activity relationships. Free Radical Biology and Medicine, 22, 749-760; (b) Rice-Evans CA, Miller NJ, Panaga G. (1996) Structure-antioxidant activity relationships of flavonoids and phenolic acids. Free Radical Biology and Medicine, 20, 933-956; (c) Lien EJ, Ren S, Bui H, Wang R. (1999) Quantitative structure-activity relationship analysis of phenolic antioxidants. Free Radical Biology and Medicine, 26, 285-294; (d) Montoro P, Braca A, Pizza C, De Tommasi N. (2005) Structure-antioxidant activity relationships of flavonoids isolated from different plant species. Food Chemistry, 92, 349-355. ICH Q2B, International Conference on Harmonisation, London, 1995. (a) Li X, Xiong Z, Ying X, Cui L, Zhu W, Li F. (2006) A rapid ultra-performance liquid chromatography-electrospray ionization tandem mass spectrometric method for the qualitative and quantitative analysis of the constituents of the flower of Trollius ledibouri Reichb. Analytica Chimica Acta, 580, 170-180; (b) Benavides A, Montoro P, Bassarello C, Piacente S, Pizza C. (2006) Catechin derivatives in Jatropha macrantha stems: Characterisation and LC/ESI/MS/MS quali-quantitative analysis. Journal of Pharmaceutical and Biomedical Analysis, 40, 639-647; (c) Montoro P, Tuberoso CIG, Perrone A, Piacente S, Cabras P, Pizza C. (2006) Characterisation by liquid chromatography -electrospray tandem mass spectrometry of anthocyanins in extracts of Myrtus communis L. berries used for the preparation of myrtle liqueur. Journal of Chromatography A, 1112, 232-240. Merfort I, Wendisch D. (1992) New flavonoid glycosides from Arnicae flos DAB 9. Planta Medica, 58, 355-357. Grouiller A, Pacheco H. (1967) Flavonoid compounds. VI. Nuclear magnetic resonance spectra of some O-glucosylflavonals, their aglycons and three synthetic mono- and di- O –glucosylflavanones. Bulletin de la Societe Chimique de France, 6, 1938-1943. Nawwar MAM, El-Mousallamy AMD, Barakat HH. (1989) Quercetin 3-glycosides from the leaves of Solanum nigrum. Phytochemistry, 28, 1755-1757. Senatore F, D'Agostino M, Dini I. (2000) Flavonoid glycosides of Barbarea vulgaris L. (Brassicaceae). Journal of Agricultural and Food Chemistry, 48, 2659-2662. NPC Natural Product Communications HPLC/DAD/ESI-MS Analysis of Non-volatile Constituents of Three Brazilian Chemotypes of Lippia alba (Mill.) N. E. Brown 2008 Vol. 3 No. 12 2017 - 2020 Patrícia Timóteoa*, Anastasia Kariotia, Suzana G. Leitãob, Franco Francesco Vincieri a and Anna Rita Biliaa a Department of Pharmaceutical Sciences, University of Florence, Via Ugo Schiff, 6, 50019, Sesto Fiorentino (FI), Italy b Faculty of Pharmacy, Federal University of Rio de Janeiro, Bloco A, 2° andar, Ilha do Fundão, 21941-590, Rio de Janeiro, Brazil patimoteo@gmail.com Received: August 27th, 2008; Accepted: November 3rd, 2008 Aqueous preparations and ethanolic extracts of three Brazilian chemotypes of Lippia alba (Mill.) N.E. Brown (Verbenaceae) were investigated for the chemical variability of their non volatile constituents by HPLC/DAD/ESI-MS analysis. The main class of compounds in all the extracts investigated was phenylpropanoids, mainly verbascoside, followed by the flavonoids tricin-7-O-diglucuronide (present in Lippia alba chemotypes 2 and 3), luteolin-7-O-glucuronide (present in L. alba chemotype 1) and mono-and di-O-glucuronic derivatives of apigenin and tricin. Four iridoids, geniposidic acid, theveside, 8-epi-loganin and mussaenoside were also identified. Keywords: Lippia alba, Verbenaceae, Brazilian chemotypes, flavonoid glycosides, phenylpropanoid glycosides, iridoids. Lippia alba (Mill.) N. E. Brown (Verbenaceae), is a very common herb in Brazil, where is popularly known as ‘Erva-cidreira’. It occurs in all regions of the country as a spontaneous or cultivated plant. In Brazilian folk medicine infusions and decoctions of its leaves are traditionally used as a sedative and for gastrointestinal disorders. Ethanolic preparations are also popularly used for fever, coughs and asthma [1,2]. There are approximately eighteen chemotypes of L. alba, mainly based on the composition of their essential oil. Brazilian chemotypes have been classified by Matos and coworkers according to the percentage of citral (chemotype 1), carvone (chemotype 2) and linalool (chemotype 3). However, the last is hardly found in Brazil as a spontaneous plant [3,4]. Several studies have been carried out regarding the characterization of volatile compounds, but to the best of our knowledge nothing has been reported yet concerning the distribution of non-volatile constituents by chemotypes, which would be useful for pharmacological purposes. Since the chemical variability of the three Brazilian chemotypes seems to be important for both the volatile and non-volatile constituents, the present study aimed to investigate the polar extracts of these chemotypes in order to establish differences between them. Infusions, decoctions and ethanolic extracts of the leaves of three chemotypes of L. alba were investigated according to their traditional uses [4,5]. The samples were submitted to HPLC/DAD/ESI-MS analyses in order to obtain a complete characterization of these preparations. In Fig. 1 the HPLC/DAD profiles of the infusions of the three chemotypes of L. alba at different wavelengths are presented: 240 nm (monitoring of iridoids), 330 nm (monitoring of phenylpropanoids) and 350 nm (monitoring of flavonoids). Results are shown in Table 1, where the identified constituents in the extracts of the three chemotypes are presented. 2018 Natural Product Communications Vol. 3 (12) 2008 Timóteo et al. Table 1: Identified compounds in the three extracts of the Lippia alba compared by chemotypes. Cpds 1 2 3 4 5 6 7 8 9 10 11 12 13 LA1I + + + t t t + + + + + + LA2I LA3I LA1D LA2D LA3D LA1E LA2E LA3E + + + + + + + + + + + + + + + + + t + t t t t t t + t + + t + + t t t t t t + + + + + + + + + + + + + + + + + + + + + + + + + t t t - Cpds: compounds; LA: Lippia alba; 1, 2, 3: chemotypes: 1 (citral), 2 (carvone), 3 (linalool); I: infusion; D: decoction; E: ethanolic extract. Iridoid compounds: (1) theveside; (2) geniposidic acid; (3) 8-epi-loganin; (4) mussaenoside; Flavonoid derivatives: (5) apigenin-7-O-glucuronide; (6) apigenin-7-O-diglucuronide; (7) tricin-7-O-diglucuronide; (8) tricin-7O-glucuronide: (9) luteolin-7-O-glucuronide; Phenylpropanoid derivatives: (10) calceolarioside E; (11) verbascoside; (12) isoverbascoside; (13) β-OH-acteoside diastereisomers; t: traces; + presence of the compound; -: absence of compound. The constituents of the extracts from these chemotypes belong mainly to three classes of compounds: iridoids, flavonoids and phenylpropanoids. This finding is in good agreement with literature data [5]. Four iridoids, theveside (1), geniposidic acid (2), 8-epi-loganin (3) and mussaenoside (4), were detected in infusions and decoctions of all chemotypes studied, in accord with literature data [6-8]. Concerning the phenylpropanoid content of the samples, four phenylpropanoids were detected, namely, calceolarioside E (10), verbascoside (11), isoverbascoside (12) (isobaric isomer of verbascoside), and a pair of diastereoisomeric forms of β-OH-acteoside (13). Compound 13 was detected in infusions and decoctions, but not in the ethanolic extracts. Phenylpropanoids 10-12 have been previously reported in the literature for L. alba whereas β-OH-acteoside was detected for the first Table 2: Positive and negative MS fragmentation and Uv-vis absorption data of the compounds detected in the three chemotypes of Lippia alba. tR (min) UV-vis Λmax (MeOH) eV MW ESI-MS+ m/z (rel. intensity, %) ESI-MS- m/z (rel. intensity, %) 3.8 236 80 390 413 (100) [M+Na+]+; 429 (20) [M+K+] + 389 (100) [M-H]-; 227 (20) [Aglycone-H]- 4.4 238 80 390 413 (72) [M+Na+]+; 429 (24) [M+K+] + 389 (100) [M-H]-; 227 (14) [Aglycone-H]- Geniposidic acid (3) 4.8 234 80 374 397 (100) [M+Na+]+; 413 (25) [M+K+] +; 787 (5) [M+K+] + 373 (73) [M-H]-; 211 (30) [Aglycone-H]- Mussaenoside (4) 12.6 238 80 + + + + 390 413 (50) [M+Na ] ; 429 (36) [M+K ] ; 229 (20) [Aglycone +H]+ Apigenin 7-O-glucuronide (5) 25.1 342 180 446 447 (100) [M+H]+; 271 (12) [Aglycone+H]+ 445 (35) [M-H]-; 285 (100) [Aglycone-H]- Apigenin 7-O- diglucuronide (6) 18.4 350 180 622 623 (100) [M+H]+ 621 (45) [M-H]-; 269 (38) [Aglycone-H]-; 351 (100) [M-H-Aglycone]- or [(2x glucuronic acid)-H]- Tricin 7-O-diglucuronide (7) 19.4 350 180 682 683 (100) [M+H]+ 681 (78) [M-H]-; 329 (12) [Aglycone-H]-; 351 (100) [(2x glucuronic acid)-H]- Tricin 7-O-glucuronide (8) 25.6 348 180 506 507 (100) [M+H]+; 331 (10) [Aglycone]+ 505 (100) [M-Hx]-; 329 (12) [Aglycone-H]-; 351 (100) [(2 glucuronic acid)-H]- 21.8 350 180 462 463 (100) [M+H]+ 461 (35) [M-H]-; 285 (100) [Aglycone-H]- 21.2 330 180 610 - 609 (100) [M-H]-; 447 (20) [M-H-162]- Verbascoside (11) 22.3 330 180 624 - 623 (100) [M-H]-; 461 (9) [M-H-caffeic acid]-; 161 (20) [caffeic- acid-H2O-H]- Isoverbascoside (12) 23.9 330 180 624 - 623 (100) [M-H]-; 461 (9) [M-H-caffeic acid]-; 161 (20) [caffeic- acid-H2O-H]- 16.7/17.3 330 180 640 - Compound Theveside (1) 8-epi-loganin (2) Luteolin 7-O-glucuronide (9) Calceolarioside E (10) β-OH-acteoside diastereoisomers (13) 389 (100) [M-H]-; 227 (18) [Aglycone-H]- 639 (100) [M-H]-; 621 (30) [M-H-H2O]-; 459 (16) [M-H-caffeic acid- H2O]-; 179 (20) [caffeic acid-H]- Constituents of three Brazilian chemotypes of Lippia alba mAU 240 nm 120 100 80 60 40 20 0 1 3 0 mAU 30 25 20 15 10 5 0 -5 Infusion Lippia alba chemotype I: LA1I 2 5 4 10 15 20 330 nm 13 13 0 mAU 20 15 10 5 0 -5 5 0 15 15 20 9 5 240 nm 40 30 20 10 0 10 2 1 mAU 40 30 20 10 0 mAU 0 15 25 30 min 5 8 20 25 5 15 20 25 30 min 11 13 13 5 10 10 15 20 25 6 10 15 30 min 5 8 20 25 30 min 240nm Infusion Lippia alba chemotype III: LA3I mAU 2 70 60 50 40 30 20 10 0 1 1 2 50 40 30 20 10 0 350nm 5 1 1 20 6 0 5 10 15 2 30 min 11 12 10 13 13 0 mAU 1 330nm 40 30 20 10 0 To the best of our knowledge, this is the first time that β-OH-verbascoside (13) has been found in L. alba, and tricin-7-O-diglucuronide (7) in the Lippia genus. For each chemotype, only small differences were observed among the three preparations. However, differences were detected between the chemotypes, especially regarding the flavonoids (Figure 1). These differences should be taken into consideration when pharmacological studies are carried out. 4 3 0 mAU Table 2 reports the UV-vis absorptions and MS fragmentation profiles of all the compounds detected in L. alba infusions, decoctions and ethanolic extracts. 12 7 5 In addition, a pair of diastereoisomeric forms of β-OH-acteoside (13) was detected. These are an analogue of verbascoside with a hydroxyl group in the β position: 3,4-dihydroxyl-phenyl-ethanol moiety. min 4 10 330 nm 0 30 11 350 nm 60 50 40 30 20 10 0 min Infusion Lippia alba chemotype II: LA2I 3 0 30 12 10 350 nm mAU 25 11 Natural Product Communications Vol. 3 (12) 2008 2019 25 30 min 7 5 8 20 25 30 Figure 1: Chromatographic profiles at 240, 330 and 350 nm of a representative samples (infusions) of the three chemotypes of Lippia alba considered. Compound 7 was only detected in chemotypes 2 and 3, and compound 9 in chemotype 1. time in this plant. Its presence was confirmed by, comparing UV, MS and retention time data with those reported previously for this compound [9,10]. Finally, five flavonoids, namely apigenin-7-Omonoglucuronide (5), apigenin-7-O-diglucuronide (5); tricin-7-O-monoglucuronide (8) tricin-7-Odiglucuronide (7) and luteolin-7-O-glucuronide (9), were detected, in accordance with literature data [11,12]. As shown in Table 1, and as expected, the main class of compounds of L. alba was phenylpropanoids, represented mainly by verbascoside (11), followed by the flavonoids tricin-7-O-diglucuronide (7) and luteolin-7-O-glucuronide (9). From the results described above, the flavonoids 7 and 9 could be used as markers to distinguish the three Brazilian chemotypes of L. alba, since 7 seems to be present only in chemotypes 2 and 3, and compound 9, only in chemotype 1. Experimental Chemicals: All solvents used were HPLC grade; CH3CN and MeOH for HPLC were purchased from Merck (Darmstadt, Germany). Formic acid (85 %) was provided by Carlo Erba (Milan, Italy). Water was purified by a Milli-Qplus system from Millipore (Milford, MA, USA). A 0.45 mm PTFE membrane filter was purchased from Waters Co. (Milford, MA). Plant material: Dried leaves of three cultivated chemotypes of L. alba (Mill.) N. E. Brown, were collected in 2008 from the Herbarium CESJ (Federal University of Juiz de Fora), MG, Brazil. Herbal preparations: Infusions: Dried powered leaves of each chemotype of L. alba (1 g) were extracted with 20 mL of boiling water. The mixture was cooled for 20 min and then filtered. Decoctions: Dried powered leaves of three chemotypes of the plant (1 g) were put in 20 mL of water and both were boiled for 2 min before filtration. Ethanolic extracts: Dried powered leaves of each chemotype of L. alba (1 g) were extracted three times successively with EtOH for 24 h each time. All preparations were lyophilized and then freeze-dried. For HPLC-DADMS analysis, the samples were obtained by dissolving and filtering the solid residues (1 mg exactly weighed) in 1 mL of MeOH. 2020 Natural Product Communications Vol. 3 (12) 2008 General experimental procedures: HPLC/DAD/ESIMS analysis was performed on a HP 1100 L instrument with DAD and managed by a HP 9000 workstation interfaced with a HP 1100 MSD APIUSA. The column used was a Varian Polaris TM C18-E (250 x 4.6 mm i.d., 5 μm maintained at 26°C. Eluents were H2O adjusted to pH 3.2 with formic acid (A), and acetonitrile (B). A multi-step linear gradient was applied from 87% A to 85% in 10 min; in 10 min to 75% B and a plateau for 3 min; 2 min to 95% CH3CN and a final plateau for 3 min. Total time analysis was 28 min, and equilibration time 10 min. Flow rate: 0.8 mL min -1. Oven temperature: 26°C. UV-vis spectra were registered between 220-500 nm and the chromatographic profiles were registered at 240, 330 and 350 nm. Mass spectrometry conditions were optimized in order to achieve sensitive values: negative and positive ionisation mode, scan spectra from m/z 100 to 800, gas temperature: 350°C, Timóteo et al. nitrogen flow rate: 10L min-1, nebulizer pressure 30 psi, quadrupole temperature: 30°C, capillary voltage: 3500 V. Applied fragmentors range: 80-180 V. Identification of constituents was carried out by HPLC/DAD/ESI-MS analysis. UV-vis and mass spectra of the peaks were compared with those of authentic standards, previously isolated compounds and literature data. Acknowledgments – Supported by the Programme Alβan, the European Union Programme of High Level Scholarships for Latin America, scholarship no: (E06M104124BR). The authors would also like to thank Professor Lyderson F. Viccini and his undergraduate students (Laboratory of Genetic, Department of Biology/ICB), University of Juiz de Fora (MG, Brazil) for providing the L. alba chemotypes. References [1] Lorenzi H, Matos FJA. (2002) Plantas Medicinais no Brasil: Nativase Exóticas Cultivadas. Instituto Plantarum (Ed.) São Paulo, Brazil, 488-489. [2] Matos FJA. (1996) As ervas cidreiras do Nordeste do Brasil. Estudo de três quimiotipos de Lippia alba (Mill.) N.E.Brown (Verbenaceae) Parte II – Farmacoquimica. Revista Brasileira de Farmácia, 77, 137-141. [3] Tavares ES, Julião LS, Lopes L, Bizzo HR, Lage CLS, Leitão SG. (2005). Análise do óleo essencial de folhas de três quimiotipos de Lippia alba (Mill.) N. E. Brown (Verbenaceae) cultivados em condições semelhantes. Revista Brasileira de Farmacognosia. 15, 1-15. [4] Gilbert B, Ferreira JP, Alves LF. (2005) Monografias de Plantas Medicinais Brasileiras e Aclimatadas. Abifito (Ed), Curitiba, 61-77. [5] Hennebelle T, Sahpaz S, Joseph H, Bailleul F. (2008) Ethnopharmacology of Lippia alba. Journal of Ethnopharmacology, 116, 211-222. [6] Hennebelle T, Sahpaz Joseph H, Bailleul F. (2006) Phenolics and iridoids of Lippia alba. Natural Product Communications, 1, 727-730. [7] Alipieva K, Kokubun T, Taskova R, Evstatieva L, Handjieva N. (2007) LC-ESI-MS analysis of iridoid glycosides in Lamium species. Biochemical Systematics and Ecology, 35, 17-22. [8] Filho JGS, Duringer JM, Uchoa, DEA, Xavier HS, Filho JMB, Filho, RB. (2007) Distribution of iridoid glycosides in plants from the genus Lippia (Verbenaceae): An investigation of Lippia alba (Mill.) N.E. Brown. Natural Product Communications, 2, 715-716. [9] Bilia AR, Giomi M, Innocenti M, Gallori S, Vincieri FF. (2008) HPLC-DAD-ESI-MS analysis of the constituents of aqueous preparations of verbena and lemon verbena and evaluation of the antioxidant activity Journal of Pharmaceutical and Biomedical Analysis, 46, 463-470. [10] Li L, Tsao R, Yang R, Liu C, Young JC, Zhu H. (2008) Isolation and purification of phenylethanoid glycosides from Cistanche deserticola by high-speed counter-current chromatography. Food Chemistry, 108, 702-710. [11] Kowalska I, Stochmal A, Kapusta I, Janda B, Pizza C, Piacente S, Oleszek W. (2007) Flavonoids from barrel medic (Medicago truncatula) aerial parts. Journal of Agricultural and Food Chemistry, 55, 2645-2652. [12] Stochmal A, Simonet AM, Macias FA, Oleszek W. (2001) Alfalfa (Medicago sativa L.) flavonoids. 2. Tricin and chrysoeriol glycosides from aerial parts. Journal of Agricultural and Food Chemistry, 49, 5310-5314. NPC Natural Product Communications Optimization and Validation of an HPLC–Method for Quality Control of Pueraria lobata Root 2008 Vol. 3 No. 12 2021 - 2027 Lidiya Bebrevska, Mart Theunis, Arnold Vlietinck, Luc Pieters and Sandra Apers* Laboratory of Pharmacognosy and Pharmaceutical Analysis, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium sandra.apers@ua.ac.be Received: June 25th, 2008; Accepted: October 19th, 2008 Pueraria lobata Willd. (Fabaceae) is a widely used medicinal plant, known as “Kudzu” and “Ge” in Japanese and Chinese traditional medicine, respectively. P. lobata is a rich source of isoflavones with phytoestrogenic properties, and its commercial use is widespread. In this study the optimization and validation of an HPLC-method for quality control of Pueraria root material is presented. By means of this analytical method the major individual constituents, i.e. the isoflavone 8-C-glycosides hydroxypuerarin, puerarin, methoxypuerarin and xylosylpuerarin, the 7-O-glycosides daidzin and genistin, and the aglycones daidzein and genistein could be quantified. The extraction procedure and the extraction solvent composition were optimized in order to ensure the exhaustive extraction of the plant material. The HPLC - conditions were evaluated and optimized for the exact quantification of all individual compounds. The HPLC analysis was carried out on a Agilent XDB RP C18 (150 x 4.6) column eluted with a binary system consisting of water (+ 0.01% formic acid) and methanol (+ 0.01% formic acid) using a linear gradient; detection was at 262 nm. Daidzin, daidzein, genistin and genistein were used as external standards. Due to the great difference in content between the C-glycosides on the one hand, and daidzin, genistin and the aglycones on the other, two separate HPLC runs were necessary for a complete analysis. The final method was fully validated according to the ICH guidelines in terms of linearity, precision and accuracy. Linear relationships for the responses of all four standards were proven. The method was shown to be repeatable for the individual compounds, i.e. RSD% between days values were within 2 to 7.5%. The accuracy of the method was demonstrated to be equal to 100% by recovery experiments. These validation results demonstrated the suitability of the method for the precise and accurate determination of Pueraria root isoflavones. Keywords: HPLC, quality control, method validation, Pueraria lobata root, isoflavones, phytoestrogenes. Pueraria lobata (Willd.) Ohwi (Fabaceae) is a medicinal plant widely used in the Oriental systems of traditional medicine. It is known as “Kudzu” in Japanese and as “Ge” in Chinese traditional medicine. It is recommended for the alleviation of different conditions and the curative properties of this drug are known to be due to its isoflavonoid phytoestrogen content. The major isoflavonoids present in Pueraria root are the 8-C-glycosides puerarin, hydroxypuerarin, methoxypuerarin and xylosylpuerarin, the 7-O-glycosides daidzin and genistin, and the aglycones daidzein and genistein (Fig. 1). Numerous commercial preparations of isoflavone extracts from the root of the plant are marketed as dietary supplements with different health promoting properties. Great attention has been paid to the analysis of isoflavones because of their importance to human health, especially in age-related and hormone-dependent diseases, such as cancer, cardiovascular diseases, menopausal symptoms, and osteoporosis [1]. Several reports have been published on the determination of Pueraria isoflavones in different matrices, i.e. Pueraria plant preparations, blood and urine samples, using HPLC or capillary electrophoresis, but these methods have yielded variable recoveries and/or were designed to determine only a limited number of phytoestrogens [2-4]. A narrow-bore HPLC-MS method was developed for the identification of Pueraria root constituents, but did not provide sufficient resolution for their quantification in routine LC-UV analysis [5]. Another method reported for the quantification of isoflavones in Kudzu root extracts suffered from poor 2022 Natural Product Communications Vol. 3 (12) 2008 R1 R5 4 5 6 O O 3 7 O R2 3’-Hydroxypuerarin Puerarin 3’-Methoxypuerarin 6”-O-Dxylosylpuerarin Daidzin Genistin Daidzein Genistein O 8 R4 2 1 R3 R1 H H H H R2 H H H H H OH H OH Glc Glc H H R3 Glc Glc Glc GlcXyl H H H H R4 OH H OCH3 H R5 H H H H H H H H H H H H Figure 1: Structure of isoflavonoid glycosides and aglycones from Pueraria root. resolution, and only puerarin was quantified [6]. In the work of Delmonte et.al., the isoflavone content was determined in dietary supplements containing soy, red clover and Kudzu, but since the glycosides were hydrolyzed during sample preparation the real composition was not determined [7]. The method developed in our laboratory to determine the isoflavone content in soy extracts could not be applied directly since here we need to extract the isoflavones from the root material.[8]. The methods available for quantitative determination of Pueraria root isoflavones in plant material or dietary supplements are limited to the detection of one or only a limited number of compounds, including the official method in the current edition of the Chinese Pharmacopoeia, which only determines the puerarin content [9,10]. Several recent papers dealing with the use of LC-MS for the analysis of isoflavones in crude mixtures have recently being published [11-15]. The aim of these studies was to identify isoflavone aglycones and major glycosides in Pueraria root by using LC-MS-MS with different modes of MS, for example neutral loss-scan mass spectrometry, selected ion monitoring (SIM), and selected reaction monitoring (SRM). From this short review of the literature it appears that no fully optimized and validated method quantifying all major constituents for routine quality control of Pueraria root material or its dry extract is available. Recently we have published the development and validation of an HPLC method for quality control of Pueraria lobata flower [16]. However, the main constituents of the flower, i.e. the isoflavone tectorigenin and two of its glycosides, were completely different from the root constituents. When developing a quantitative method, it must be Bebrevska et al. considered that the therapeutic potency of a preparation depends not only on the amount of the active ingredients, but also on their absorption profile. The pharmacokinetic profiles of the C- and O-glycosides present in Pueraria root are different, and therefore special attention should be paid to their individual determination. The method presented here was designed to determine separately a maximum number of compounds, i.e. all major isoflavones mentioned above. In our samples of P. lobata root material 3’hydroxypuerarin, puerarin, 3’-methoxypuerarin, 6”xylosylpuerarin, daidzin, genistin, daidzein and genistein could be identified. Puerarin, 3’-methoxypuerarin, daidzin and daidzein were isolated, and identified by NMR spectroscopy (1H NMR, 13C NMR, DEPT-135 and DEPT-90, and 2D NMR when necessary, including 1H-1H DQF-COSY, HSQC and HMBC) and mass spectrometry. The NMR assignments were in agreement with literature data (Supplementary data: Table 1S) [17-19]. The presence of the other constituents in the root extract was confirmed by LC-MS analysis, by comparison with published data and standards of genistein and genistin. For the development of an analytical method, first the separation conditions were optimized in order to achieve satisfactory resolution. Because of the complexity of the mixture this was only possible when using rather long gradient conditions and run times. The initial set of separation conditions is summarized in Table 1. The LiChrosphere column did not provide the desired separation of the major peaks with any of the tested eluents or gradients, which included water / methanol mixtures without and with addition of 0.01% formic acid (pH ≈ 3 – 4). Therefore, the separation on a Zorbax Eclipse XDB column was tested in combination with a linear gradient of a binary phase: water + 0.01% formic acid and methanol + 0.01% formic acid. These conditions provided satisfying resolution of the peaks (Figure 2, Table 2). The obvious difference in selectivity between the two column packings might be due to differences in the amount of bonded phase per unit surface of the silica particles (density), the way in which the bonded phase is attached to the silica surface, and the purity of the silica. Another problem that had to be overcome in the design of the procedure was the big difference in content between the glycosides and the aglycones. If Quality control of Pueraria lobata root Table 1: Initial separation conditions. Separation variable Column Length x ID (mm) Particle size (µm) Stationary phase Mobile phase Detection Flow rate (ml/min) Injection volume standards (µl) Solution LiChrosphere 250 x 4 5 C 18 5% methanol in water to 100% for gradient elution over 60 min (broad linear gradient to survey the mixture) 262 nm 1.0 20 / 100 Sample volume (µl) Standard solutions 20 / 100 20/100 Table 2: Final separation conditions. Separation variable Column Length x ID (mm) Particle size (µm) Stationary phase Mobile phase Gradient time (min) Wash up and equilibration (min) Run time (min) Detection Flow rate (ml/min) Standard solutions volume Sample volume (µl) Solution Agilent Zorbax Eclipse XDB 150 x 4.6 5.0 C 18 XDB 15% methanol + 0.01% formic acid to 65% methanol + 0.01% formic acid for gradient elution 50 10 60 262 nm 1.0 20 / 100 20 / 100 Figure 2: HPLC profile of Pueraria root extract. The glycosides elute between 5 and 20 min and the aglycones start from 22 min. 20 µL of the final solution was injected, the aglycones were barely detectable, therefore it was necessary to measure them in a second separate run with an injection volume of 100 µL. All peaks of interest were separated from the adjacent ones allowing their exact integration. The peaks of hydroxypuerarin (peak 5 on the chromatogram), genistin (13), daidzin (11), daidzein (18) and genistein (21) were well separated from their neighboring peaks. The critical band couples were: puerarin (peak 8 on the chromatogram) / 3’-methoxypuerarin (9), 3’-methoxypuerarin (9) / 6”xylosylpuerarin (10), and 6”-xylosylpuerarin (10) / daidzin (11). However, they were successfully resolved for the purpose of quantification. All peaks of interest are symmetric. Natural Product Communications Vol. 3 (12) 2008 2023 The extraction procedure and the extraction solvent composition were optimized in order to ensure the exhaustive extraction of the plant material, in the same way as described for Pueraria flower [16]. The final conditions are described in the experimental section. Daidzin, daidzein, genistin and genistein were chosen as external standards. The content of 3’-hydroxypuerarin, puerarin, 3’-methoxypuerarin, 6”-xylosylpuerarin and daidzin were calculated with respect to daidzin, while the contents of genistin, daidzein and genistein were calculated with respect to each individual standard. The standards were analyzed using the same HPLC conditions as for the analytes. This method was validated according to the ICH guidelines on the validation of analytical methods [20,21]. The calibration functions for each of the four standards were investigated by injecting six different concentrations of reference solution in duplicate. The concentration level for the isoflavone glycoside daidzin was within the range of 4.13 – 103.2 µg/mL, and for the glycoside genistin and the aglycones daidzein and genistein within the range of 0.20 – 4.0 µg/mL, 0.43 – 8.68 µg/mL, and 0.034 – 0.54 µg/mL, respectively. In order to describe and investigate the relationship, least square lines and the correlation coefficients were calculated. The significance of the slopes (a≠0) and the intercepts (b=0) of the obtained functions were tested by means of Student’s t-test. To check whether the linear model fitted the obtained data a lack-of-fit (LOF) test was performed and the resulting residuals were graphically inspected. The response functions of the four standards were investigated and the results are shown in Table 3. Graphical inspection of the residuals, the LOF test and the correlation coefficients proved that a linear model response function could be fit to the data in the inspected range. The t-test of the intercept revealed that point (0.0) fell within each of the calibration curves. This implied that for routine analysis in this range, the isoflavone content could be determined against a single standard concentration level instead of constructing a calibration line each time the analysis is performed. The repeatability and the time-different intermediate precision were evaluated by analyzing six independently prepared samples (100% level, i.e. performed on 0.600g of plant material) on three 2024 Natural Product Communications Vol. 3 (12) 2008 Bebrevska et al. Table 3: Overview of response function data for the reference materials used. Concentration range (µg/mL) Number of levels (n=2) Analysis of the residuals Correlation coefficient 95% CI of the intercept Intercept ± standard error Slope ± standard error ANOVA Lack of fit, F=MSLOF/MSPE (Fcrit = 5.409) Daidzin 4.13 – 103.2 6 Randomly scattered 0.9995 [-53879 +23646] -15117 ± 17397 31747 ± 303 Genistin 0.434 – 8.68 6 Randomly scattered 0.9999 [- 751 +10261 ] 4755 ± 2471 209128 ± 586 Daidzein 0.2012 – 4.024 6 Randomly scattered 0.9998 [-746 +7047] 3150 ± 1749 217842 ± 895 Genistein 0.0335 – 0.5372 6 Randomly scattered 0.9992 [-3317 +1119] -1099 ± 995 292237 ± 3743 2.1 1.0 2.3 4.8 Table 4: Overview of the precision data. Parameter Repeatability / Time-intermediate precision Number of replicates on each day Number of days Mean content (%) Day 1 Day 2 Day 3 RSD (%) Day 1 Day 2 Day 3 RSD% between days < 5% Linearity / Intermediate precision on different conc. levels Numbers of replicates on each level Number of levels Mean content (%) 50 200 RSD (%) 50 200 Cochran’s test (C critical = 0.506) RSD% between levels (%) < 5% Hydroxypuerarin Puerarin Methoxypuerarin Xylosylpuerarin Daidzin Genistin Daidzein Genistein 6 6 6 6 6 6 6 6 3 3 3 3 3 3 3 3 0.86 0.83 0.83 3.06 3.06 2.95 0.80 0.87 0.84 0.67 0.64 0.61 0.58 0.63 0.59 0.11 0.11 012 0.074 0.071 0.067 0.0074 0.0082 0.0074 1.09 0.60 1.47 1.16 0.64 0.78 1.64 0.87 0.83 1.09 0.93 0.81 1.18 0.35 0.82 5.26 2.56 6.10 3.59 2.50 3.60 4.19 5.87 4.29 2.21 2.34 4.30 4.91 5.51 7.07 5.05 7.29 6 6 6 6 6 6 6 6 3 3 3 3 3 3 3 3 0.84 0.80 3.21 3.08 0.83 0.83 0.65 0.62 0.60 0.60 0.11 0.11 0.076 0.068 0.0094 0.0051 1.04 0.62 0.334 1.26 0.34 0.417 2.31 1.44 0.464 1.21 0.64 0.305 1.62 1.63 0.360 4.15 4.73 0.355 4.54 1.30 0.410 3.02 1.82 0.446 4.15 3.18 3.22 4.03 3.43 6.13 5.98 21.46 different days. The mean content and the betweendays RSD (%) for each compound are presented in Table 4. The mean, the standard deviation and the relative standard deviation were calculated for each day. ANOVA single factor was used to compare the results of the three days. Within- and between-days relative standard deviations were calculated. To test the precision of the method in terms of between concentration levels variation, six samples weighing twice the normal weight (200%) and six samples weighing half the normal weight (50%) were analyzed following the same procedure. The mean, the standard deviation and relative standard deviations were calculated for each level. By means of a Cochran’s test (5 groups, n=6), the homogeneity of the variances of experiments at the three concentration levels (50, 100 (3 days), and 200%) was determined prior to ANOVA single factor comparison of the results for the three levels. Withinand between-level relative standard deviations were calculated. The major isoflavone was the C-glycoside puerarin present in an amount of 3.03%, followed by 3’-hydroxypuerarin 0.84%, 3’-methoxypuerarin 0.83%, 6”-xylosylpuerarin 0.64%, and daidzin 0.60%. Genistin and daidzein were present in minor amounts of 0.11% and 0.07%, respectively. Genistein was present at a trace level of 0.008%. The precision of the method was acceptable for the major compounds 3’-hydroxypuerarin, puerarin, 3’-methoxypuerarin, 6”-xylosylpuerarin and daidzin with an RSD (%) of 2.21, 2.34, 4.30, 4.91 and 5.51% respectively. The RSD between-days (%) values for genistin, daidzein and genistein were 7.07, 5.05, and Quality control of Pueraria lobata root Natural Product Communications Vol. 3 (12) 2008 2025 7.29%, respectively. They are also acceptable with respect to the low amount in which these isoflavones are present. Standards of daidzin (100 mg, purity 99.2%), daidzein (25 mg, purity 96.7%), genistin (100 mg, purity 99.1%) and genistein (50mg, purity 100%) were purchased from LC Laboratories (Woburn, Massachusetts, USA). The precision of the measurements in the range between 50% and 200% of the expected amount of each analyte was investigated. The results are also presented in Table 4. The variances of the measurements for the three levels were compared by means of Cochran’s test. Since for each of the isoflavones the calculated C-values were lower than the critical value, the precision of the determinations within this range can be considered equal. The RDS between-levels (%) were still acceptable and no trend, i.e. lower values at 200% and higher values at 50%, was detected. The accuracy of the method was investigated by performing recovery experiments according to the method of standard additions (Supplementary data, Tables 2S – 5S). Daidzin, genistin, daidzein and genistein stock solutions were added before the extraction at different concentration levels to half of the normally analyzed amount of plant material (300 mg). At each level the samples were prepared in triplicate and analyzed. Mean recoveries of 99.05% (RSD% = 3.21), 101.55% (RSD% = 4.32), 99.16% (RSD% = 1.92), and 105.56% (RSD% = 6.36) were determined for daidzin, genistin, daidzein and genistein, respectively. The mean recoveries (%) were verified not to be significantly different from 100 % for all four isoflavones by means of Student’s t-test, which proved that our method produces accurate results. These results proved that the reported method correctly quantifies all major isoflavones present in Pueraria root with acceptable accuracy and precision. This study proved the suitability of the method for the quality control of Pueraria root crude drug. Experimental Materials: Distilled water (RiOs) was obtained from a Millipore water purification system (Millipore, Brussels, Belgium). Methanol (HPLC-quality) and silica gel 60 (0.040-0.063) were purchased from Merck (Darmstadt, Germany), and formic acid and dimethyl sulfoxide from Acros Organics (Geel, Belgium). All solvents and reagents were of analytical grade. Preparative C18 (55-105 µm) was from Waters (Maliford, MA, USA). Dried roots of Pueraria lobata were kindly provided by SINECURA (Ghent, Belgium). The root material was investigated macro- and microscopically according to the Chinese Pharmacopoeia in order to confirm its identity. Spectroscopy: 1D NMR (1H, 13C, DEPT-135 and DEPT-90) and 2D NMR spectra (1H-1H DQF-COSY, HSQC and HMBC) were recorded in methanol-d4 on a Bruker DRX-400 instrument operating at 400.15 MHz for 1H, using standard software packages. LC-MS analysis was performed on an Agilent 1100 with a diode array detector, coupled to a Bruker Esquire 3000 plus ion trap MS (Bruker Daltonics, Billerica, USA). LC-MS analysis was carried out on a Surveyor LC system with a diode array detector coupled to a LXQ linear ion trap (Finnigan, Bremen, Germany). Thin layer chromatography: 1) Silica gel 60 F 254 plates (Merck), 20 x 20 cm, and 2) RP 18 plates (Merck), 20 x 10 cm were used. Mobile phase systems were: 1) n-butanol : ethanol : water 4 : 1 : 2.2; and 2) acetonitrile : water 1 : 1. Compounds were detected by: UV light at 254 nm; UV light at 366 nm; and spraying with p-anisaldehyde / sulfuric acid in methanol followed by heating the plate (100-105°C) for about 5 min. HPLC: The isolation of the marker compounds was carried out on an Agilent 1100 HPLC-system equipped with a DA (diode array) detector. The HPLC analyses during the method development and validation procedure were carried out using a Gilson instrument (pump model 322, UV-VIS detector model 156) equipped with a Gilson automatic injector. The analytical work was performed on an Agilent Zorbax Eclipse XDB-C18 column (150 x 4.6 mm, 5µm) and the semipreparative work on a Macherey-Nagel, Nucleosil 100-7 C18 (10 x 250 mm) semipreparative chromatographic column. Extraction and isolation: Dried roots of Pueraria lobata were pulverized and sieved in order to assure particle uniformity. A 50% ethanol extract was prepared under constant stirring by adding multiple portions of fresh extracting solvent till complete 2026 Natural Product Communications Vol. 3 (12) 2008 exhaustion of the material. The extracted portions were combined and concentrated by evaporation under reduced pressure to give the crude extract. After lyophilization, 20 g of the powder was resuspended in distilled water and partitioned between ethyl acetate and subsequently n-butanol, both saturated with water. The resulting subfractions (ethyl acetate, n-butanol and residual water) were dried under reduced pressure. The ethyl acetate fraction (5.0 g) was subjected to column chromatography on silica gel (350 g), eluted with n-butanol : ethanol : water 6 : 1 : 2.2. The 52 collected fractions were evaluated by means of TLC, and according to their profile they were combined in 7 subfractions. Subfraction 4 (3.1 g) was further fractionated on silica gel (360 g), eluted with n-butanol : ethanol : water 4 : 1 : 2.2. The resulting 18 fractions were evaluated by TLC and joined into 5 fractions. The fourth fraction was submitted to column chromatography on silica gel (50 g). The 18 obtained fractions were put together in 3 fractions. Fraction 2 was subjected to semipreparative HPLC. An HPLC gradient consisting of methanol / 0.01% formic acid and water / 0.01% formic acid was optimized for the purpose of semi-preparative separation: from 15% methanol (t = 0 min), over 21% (t = 15 min), 25% (t = 25 min), to 95% (t = 55 min). The three major compounds isolated were identified as puerarin, 3’-methoxypuerarin, and daidzin. Subfraction 3 of the ethyl acetate fraction (250 mg) was submitted to column chromatography on silica gel (30 g) and eluted with chloroform : methanol 9 : 1. The resulting 18 fractions were combined in 3 subfractions. The second subfraction was purified by semipreparative HPLC using a gradient from 15% to 67.8% methanol over 33 min at a flow rate of 2.2 mL/min. One compound was isolated, and identified as daidzein. The presence of 3’-hydroxypuerarin, 6”-xylosylpuerarin, genistein and genistin was confirmed by LC-MS analysis, by comparison with published data and standards of genistein and genistin. Preparation of the sample solution: The powdered plant material was sieved (355 μm) to ensure particles uniformity. An exactly weighed amount of 0.600 g of plant material was placed in a round bottomed flask and 10 mL dimethyl sulfoxide was added. After 15 min, 60 mL of 50% methanol was added, and refluxed for 30 min. To separate the clear liquid from the plant material centrifugation at 3000 Bebrevska et al. rpm was performed for 15 min. A fresh portion of 60 mL 50% methanol was added to the plant material, repeating the extraction procedure. The supernatants resulting from the two extractions were joined and collected in a calibrated flask of 200 mL. The flask was filled to volume with 50% methanol. Finally 10 mL of this solution was diluted to 20 mL with water. This sample solution was used for HPLC analysis. Two separate analyses were performed for each sample: one injecting 20 µL to quantify the glycosides, and another one injecting 100 µL for the quantification of the aglycones. Preparation of the standard solutions: About 10 mg of daidzin was accurately weighed and dissolved in 25 mL of dimethyl sulfoxide. An aliquot of 2 mL of this solution was diluted to 50 mL with 50% methanol. About 10.0 mg of genistin was accurately weighed and dissolved in 25 mL of dimethyl sulfoxide. An aliquot of 0.5 mL of this solution was diluted to 100 mL with 50% methanol. About 10.0 mg of daidzein was accurately weighed and dissolved in 25 mL of dimethyl sulfoxide. An aliquot of 0.5 mL of this solution was diluted to 100 mL with 50% methanol. About 10.0 mg of genistein was accurately weighed and dissolved in 25 mL of dimethyl sulfoxide. An aliquot of 0.5 mL of this solution was diluted to 50 mL with 50 % methanol. Calculation: The percentage content (Cs %) of the compounds in the plant material was calculated using the formula: Cs % = As x Cref x 2 x 200 x 100 / (Aref x m) where As was the area of the peak of the isoflavone in the sample, Aref the area of the isoflavone in the reference solution, Cref the concentration of the reference solution, and m the weight of the drug in mg. For the purpose of exact quantification, loss on drying was determined according to Ph. Eur. Article 2.2.32.d. The water content was found to be 8.8 %. Acknowledgements - Melisana (Brussels, Belgium) and the special Fund for Research of the University of Antwerp (NOI-BOF) are acknowledged for their financial support. Quality control of Pueraria lobata root Natural Product Communications Vol. 3 (12) 2008 2027 References [1] Cos P, De Bruyne T, Apers S, Vanden Berghe D, Pieters L, Vlietinck A. (2003) Phytoestrogens: Recent developments. Planta Medica, 69, 589-599. [2] Liu S, Wang J, Liu C, Wen G, Liu Y. (1998) A study on processing of the root of Pueraria lobata (Willd.) Ohwi by HPLC. Zhongguo Zhong Yao Za Zhi, 23, 723-725. [3] Chen G, Zhang J, Ye J. (2001) Determination of puerarin, daidzin and rutin in Pueraria lobata (Willd.) Ohwi with electrochemical detection. Journal of Chromatography A, 923, 255-262. [4] Askada Y, Kawano S, Yamagishi M. (1980) High-speed liquid chromatographic analysis of drugs. XIII. Determination of daidzin in Puerariae Radix. Yakugaku Zasshi, 100, 1057-1060. [5] Rong H, De Keukekeire D, De Cooman L, Baeyens WRG, Van Der Weken G. (1998) Narrow-bore HPLC analysis of isoflavonoid aglycones and their O- and C-glycosides from Pueraria lobata. Biomedical Chromatography, 12, 170-171. [6] Benlhabib E, Baker JI, Keyer DE, Singh AK. (2004) Quantitative analysis of phytoestrogens in kudzu-root, soy and spiked serum samples by high-pressure liquid chromatography. Biomedical Chromatography, 18, 367-380. [7] Delmonte P, Perry J, Rader JI. (2006) Determination of isoflavones in dietary supplements containing soy, red clover and kudzu: Extraction followed by basic or acidic hydrolysis. Journal of Chromatography A, 1107, 59-69. [8] Apers S, Naessens T, Van Den Steen K, Cuyckens F, Claeys M, Vlietinck A. (2004) Fast high-performance liquid chromatography method for quality control of soya extracts. Journal of Chromatography A, 1038, 107-112. [9] Pharmacopoeia of The Peoples Republic of China. (2005) The Pharmacopoeia Commission of the Ministry of Health of PRC. Guangdong Science and Technology Press, Guangzhou, P.R. China, p. 230. [10] Prasain JK, Kones K, Kirk M, Wilson L, Smith-Johnson M, Weaver C, Barnes S. (2003) Profiling and quantification of isoflavonoids in kudzu dietary supplements by high-performance liquid chromatography and electrospray ionization tandem mass spectrometry. Journal of Agricultural and Food Chemistry, 51, 4213-4218. [11] Zhang Y, Xu Q, Zhang X, Chen J, Liang X, Kettrup A. (2005) High-performance liquid chromatography-tandem mass spectrometry for identification of isoflavones and description of the biotransformation of kudzu root. Analytical and Bioanalytical Chemistry, 383, 787-796. [12] Zhang Y, Chen J, Zhang C, Wu W, Liang X. (2005) Analysis of the estrogenic compounds in kudzu root by bioassay and highperformance liquid chromatography. Journal of Steroid Biochemistry and Molecular Biology, 94, 375-381. [13] Boue SM, Carter-Wientjes CH, Shih BY, Cleveland TE. (2003) Identification of flavone aglycones and glycosides in soybean pods by liquid chromatography-tandem mass spectrometry. Journal of Chromatography A, 991, 61-68. [14] Wu Q, Wang M, Simon JE. (2003) Determination of isoflavones in red clover and related species by high-performance liquid chromatography combined with ultraviolet and mass spectrometric detection. Journal of Chromatography A, 1016, 195-209. [15] Tian H, Wang H, Guan Y. (2005) Separation and identification of isoflavonoids in Pueraria lobata extracts and its preparations by reversed-phase capillary liquid chromatography coupled with electrospray ionization quadrupole time of flight mass spectrometry. Se Pu, 23, 477-481. [16] Bebrevska L, Bravo L, Vandervoort J, Pieters L, Vlietinck A, Apers S. (2007) Development and validation of an HPLC method for quality control of Pueraria lobata flower. Planta Medica, 73, 1606-1613. [17] Rong H, Stevens JF, Dienzer ML, De Cooman L, De Keukeleire D. (1998) Identification of isoflavones in the roots of Pueraria lobata. Planta Medica, 64, 620-627. [18] Kinjo J, Furushawa J, Baba J, Takeshita T, Yamasaki M, Nohara T. (1987) Studies on the constituents of Pueraria lobata III. Isoflavonoids and related compounds in the roots and the voluble stems. Chemical and Pharmaceutical Bulletin, 35, 4846-4850. [19] Ohshima Y, Okuyama T, Takahashi K, Takizawa T, Shibata S. (1998) Isolation and high performance liquid chromatography (HPLC) of isoflavonoids from the Pueraria root. Planta Medica, 64, 250-254. [20] Text on Validation of Analytical Procedures – ICH Harmonized Tripartite Guideline. (1994) ICH. [21] Validation of Analytical Procedures: Methodology – ICH Harmonized Tripartite Guideline. (1996) ICH. NPC Natural Product Communications Pharmacokinetics of Luteolin and Metabolites in Rats 2008 Vol. 3 No. 12 2029 - 2036 Sasiporn Sarawek, Hartmut Derendorf and Veronika Butterweck* Department of Pharmaceutics, College of Pharmacy, University of Florida, POBOX 100494, Gainesville, FL 32610, USA butterwk@cop.ufl.edu Received: July 21st, 2008; Accepted: October 27th, 2008 The pharmacokinetic parameters of luteolin and its glucuronide/sulfate conjugates were studied in rats after a single 50 mg/kg dose of luteolin administered as intravenous bolus or oral solution. Plasma and urine samples were enzymatically hydrolyzed to determine conjugate concentrations of luteolin. Noncompartmental analysis revealed a half-life of 8.94 h for free (unconjugated) and 4.98 h for conjugated luteolin following intravenous administration. Following oral administration, plasma concentrations of luteolin attained a maximum level of 5.5 μg/mL at 5 min and decreased to below LOQ (100 ng/mL) after 1 h. Ke could not be calculated because the elimination phase was below LOQ. The low bioavailability (F) of luteolin, 4.10% at a dose of 50 mg/kg, is presumably due to the significant first pass effect. For i.v. administration, the maximum concentration of luteolin was 23.4 μg/mL at 0 h. The plasma concentration versus time profile of luteolin was biphasic, subdivided into a distribution phase and a slow elimination phase for oral and intravenous administration. Luteolin was found to have a large volume of distribution and a high clearance. Double peaks were found after intravenous and oral administration, suggesting enterohepatic recirculation. Keywords: luteolin, conjugates, pharmacokinetics. Flavonoids are polyphenolic compounds widely distributed in plants and vegetables. They have been reported to possess numerous biological effects such as antioxidant, free-radical scavenger, antiinflammatory, antiestrogenic, and antiproliferative activities [1,2]. These effects are helpful for human health, such as in the treatment and prevention of cancer, cardiovascular disease, and other pathologies [1,2]. The flavone luteolin usually occurs in its glycosylated forms in celery, green pepper, camomile tea, and artichoke. It has been reported to have antitumorigenic [3,4], anticancer [5-8], antiproliferative [9,10], and antioxidant effects [6] and has been recognized as an inhibitor of protein kinase C [11] and xanthine oxidase [12,13]. The absorption and metabolism of luteolin have been evaluated in vitro using Caco-2 cells and microsomes obtained from liver or intestine of rats and humans [14,15]. In addition, pharmacokinetic studies in animals and humans of luteolin and luteolin-rich plants have been reported [16-19]. However, until now, in vivo investigations of the disposition, absorption, bioavailability, and metabolism of luteolin are limited. Therefore, in the present experiments, the pharmacokinetics of luteolin and its glucuronide/sulfate conjugates after oral and intravenous (i.v.) injection in rats were determined. In addition, the elimination of luteolin after oral and intravenous administration was measured in urine samples. For the present investigation, we focused on the pharmacokinetic evaluation of the aglycone luteolin because recently we demonstrated that this compound produces strong xanthine oxidase inhibition in vitro [13]. The concentration-time profiles and the pharmacokinetic parameters of free luteolin after oral and i.v. administration are presented in Table 1 and Figure 1. The plasma concentration of luteolin attained a maximum level of 5.49 μg/mL 5 min after oral administration of luteolin to rats at a dose of 50 mg/kg. The low bioavailability (F) of luteolin, 4.10% at a dose of 50 mg/kg, is presumably due to the significant first pass effect. Following 2030 Natural Product Communications Vol. 3 (12) 2008 Sarawek et al. Table 1: Pharmacokinetic (PK) parameters of unconjugated luteolin after oral and i.v. administration of luteolin at a dose 50 mg/kg. Parameter ke (1/h) Cl (L/h/kg) Vdarea (L) AUC0-last (h*μg/mL) AUC0-∞ (h*μg/mL) F (%) Luteolin oral Luteolin i.v. 5 5.49 ND ND 0 23.42 0.08 8.94 2.14 27.58 20.55 23.39 0.87 0.96 4.10 Table 2: Pharmacokinetic (PK) parameters of luteolin conjugates after oral and i.v. administration of luteolin at a dose 50 mg/kg. Parameter Tmax (h) Cmax (μg/mL) ke (1/h) Luteolin conjugates oral 0.25 5.77 Luteolin conjugates i.v. 0.08 4.31 0.10 0.14 t ½ (h) 6.57 4.98 AUC0-last (h*μg/mL) 11.49 12.83 AUC0-α (h*μg/mL) 15.68 15.26 All PK parameters presented in Tables 1 & 2 are mean values calculated by a normalized dose (50 mg/kg). AUC = area under the curve; Cmax = maximal plasma concentration; Tmax = time to reach Cmax; Ke = elimination rate constant; t ½ = elimination half life; Cl/F = clearance after oral administration; Cl = clearance; Vdarea = distribution volume; AUC0-last = area under the moment curve extrapolated to Tlast; AUC0-∞ = area under the curve extrapolated to infinity; F = oral bioavailability; ND = not determined i.v. administration, the maximum concentration of luteolin was 23.42 μg/mL at 0 h. The plasma concentration versus time profile of luteolin was biphasic, subdivided into a distribution phase and a slow elimination phase for oral and intravenous administration. The concentration-time profiles and the pharmacokinetic parameters of luteolin conjugates after oral and i.v. administration are presented in Table 2 and Figure 1. Plasma concentrations of luteolin conjugates after oral and i.v. administration of luteolin attained maximum levels of 5.77 μg/mL at 0.25 h and 4.31 μg/mL at 0.08 h, respectively, and decreased to below the LOQ at 24 h. However, the half-life for luteolin was greater than that of conjugated luteolin following i.v. injection. Double peaks were found for luteolin conjugates after oral and i.v. administration at 0.25 and 1 h, respectively, suggesting it might pass enterohepatic circulation. Urinary excretion of free luteolin and its conjugates within 24 h after oral and intravenous administration of luteolin was very low (0.98 - 4.97% of the dose), which suggests that these compounds are not primarily excreted via the urine (Table 3). Figure 1: Plasma concentration-time curves. A) Luteolin after i.v. oral administration. B) Luteolin after oral administration. C) Luteolin conjugates after i.v. administration. D) Luteolin conjugates after oral administration. Luteolin 50 mg/kg was administered to rats (n = 8-11). Error bars refers to the standard deviation of concentration data at each sampling time point. Table 3: The excretory recovery for 24 h of free luteolin and luteolin conjugates in urine after oral and i.v administration of luteolin at a dose 50 mg/kg. Treatment % Luteolin % Luteolin conjugates oral 0.9 ± 0.9 3.9 ± 0.5 i.v. 2.0 ± 0.9 4.9 ± 1.7 Data expressed as mean ± SD (n = 8-11) Free luteolin and luteolin conjugates were present in rat plasma and urine after oral and intravenous administration. However, the conjugates (glucuronides or sulfates) could not be further identified in this study since the analytical method was developed only for the parent compound. The presence of free luteolin suggested that some of it can escape intestinal and hepatic conjugation. The pharmacokinetic profiles of free luteolin and luteolin conjugates in rat plasma are presented in Figure 1. When luteolin was administered orally, the maximum concentrations of luteolin and luteolin conjugates were 5.49 and 5.77 μg/mL at 5 min and 15 min, respectively. The total concentration of luteolin in rat plasma 5 min after dosing was 9.25μg/mL. Shimoi et al. [18] reported 15.5 ± 3.8 nmol/mL (4.4 ± 1.09 μg/mL) of total luteolin in rat plasma 30 min after administration of one single dose of luteolin (50 μmol/kg, 14.3 mg/kg) in propylene glycol. In dogs, the maximum concentration of luteolin was about 450 ng/mL 3 h after a single oral Pharmacokinetic parameters of luteolin Figure 2: Fitted luteolin concentrations after Experimental points represent the means of 8-11 rats. Natural Product Communications Vol. 3 (12) 2008 2031 i.v. injection. dose of a Chrysanthemum morifolium extract (102 mg/kg containing 7.60% luteolin, 7.75 mg/kg luteolin) [16]. In humans, peak plasma concentrations of total luteolin were reached within 0.5 h with a maximum level of 156.5 ± 92.29 ng/mL after a single oral dose of an artichoke leaf extract (153.8 mg containing luteolin-7-O-glucosides; equivalent to 35.2 mg luteolin) [19]. The differences observed in these studies could be explained by the initial dose administration, the solvents used to dissolve luteolin and the source of intake flavonoids (extract versus pure compound). Figure 2 illustrates the fitted luteolin concentrations versus time profiles with the two compartment body model. It can be seen that the model describes the luteolin data very well. The AIC and SC were -11.18 and -9.59, representing a good fit. The PK parameters obtained by fitting the mean concentrations versus time profiles of luteolin concentrations after i.v. treatment are presented in Table 4. The rapid absorption of several flavonoids has been reported in previous literature. For example, in humans, the main peak appeared approximately 2.9 h after quercetin administration [20,21]. In rats, flavonoids seem to appear more rapidly. When luteolin was administered via gastric intubation, the compound was detected in rat plasma after 15 min [18], whereas quercetin (given orally) appeared after 5 min [22]. Apigenin given intraperitoneally, appeared in plasma after 30 min [23]. Based on these literature data, the presence of flavonoids in the blood occurs within a few minutes to a few hours, which correlates with our results. The low bioavailability of luteolin (F = 4.1%) and the high metabolite concentrations indicate a first pass metabolism. Absorbed luteolin undergoes biotransformation (methylation, glucuronidation or sulfation), as has been shown in previous studies. For example, in vitro experiments demonstrated that 74% of luteolin was conjugated to glucuronic acid after incubation with microsomal samples from the human intestine [14]. The most common binding sites of the molecule were the hydroxyl groups in the 3′- and 4′- position (51% and 44%) [14]. Boersma et al. [14] found three glucuronosyl conjugates of luteolin, 7-O-, 3′-O- and 4′-O-glucuronosyl luteolin, after incubation with microsomes from rats and humans. Shimoi et al. [18] investigated the absorption of luteolin by rat inverted small intestine. Luteolin was recovered in rat plasma as two metabolites, the glucuronidate or sulfate forms of the O-methylate conjugate. Only a small part of the compound remained unconjugated. Murota et al. [15] reported the uptake and transport of flavonoid aglycones by human intestinal Caco-2 cells. The flavonoids quercetin, kaempferol, luteolin and apigenin were converted to their glucuronide/sulfates by Caco-2 cells, and the level of the intact aglycone was lower than those of the glucuronide/sulfates in the basolateral solution. In addition to the first-pass metabolism, the role of efflux transporters in contributing to the low oral bioavailability of drugs has been well acknowledged [24]. Among the efflux transporters located in the intestine and the liver, the ATP-binding cassette (ABC) superfamily, including several members such as P-glycoprotein (P-gp), multidrug resistance associated proteins (MRPs), and breast cancer resistance protein (BCRP/MXR), has been well investigated for its roles in intestinal and hepatic disposition of drugs [24]. Increasing evidence has demonstrated the interactions of flavonoid aglycons and glycosides with ABC transporters. For example, ginkgo flavonols, quercetin, kaempferol, and isorhamnetin were demonstrated to be the substrates of P-gp in the Bacap37/MDR1 transfected cell model [25]. The interaction of flavonoid aglycones and glycosides with ABC transporters may greatly influence their absorption, disposition and excretion in the body, and also alter pharmacokinetic profiles of the concurrently administrated drugs acting on the same transporters, thereby probably leading to significant impacts in the therapeutic outcomes. 2032 Natural Product Communications Vol. 3 (12) 2008 However, the bioavailability of luteolin is similar to that of quercetin [15]. Chen et al. [22] reported the systemic bioavailability of quercetin and quercetin conjugates as 5.3% and 55.8%, respectively, in rats. Moreover, after oral administration of quercetin, about 93.3% of it was metabolized in the gut, with only 3.1% metabolized in the liver. Interestingly, the half-life for free luteolin was greater than that for conjugated luteolin following i.v. injection. This is unusual since in theory the metabolites should not have a shorter half-life than the parent drug. However, similar results were reported by Cave et al. [26], who also reported a longer half-life for the parent compound genistein than for the genistein metabolites after i.v. injection. Therefore, the pharmacokinetic variables reported for the conjugates should be interpreted with caution, since they describe the appearance and disposition of endogenously synthesized compounds, each of which may possess distinct kinetics [26]. It also can be suggested that the terminal half-life has not been reached yet and that by using a more sensitive assay one would find a little longer half-life of the conjugates. Thus, a more complete analysis of the metabolism of luteolin will require separation into individual metabolites and kinetic analysis of the individual compounds. Only small amounts of free luteolin and luteolin conjugates were found to be eliminated in the urine in our study. This is consistent with the observations by others. Shimoi et al. [18] reported an excretory recovery of unmodified luteolin in rat urine of 4%. Luteolin conjugates were recovered only by 1.99 ± 1.50% after intake of luteolin-7-O-glucoside [19]. Comparably, only 0.58% of apigenin was recovered in urine samples within 24 h after parsley ingestion in humans [27]. Gugler et al. [28] found that after intravenous administration of quercetin (100 mg), 7.4% of the dose was excreted in the urine as a conjugated metabolite and 0.65% of the dose was recovered in the form of unchanged quercetin. Therefore, it seems that the urinary elimination of luteolin is not the main excretion route in rats. At present, it can be speculated that excretion via feces may be the main route of elimination of luteolin and, in particular, its metabolites [29]. Since in the present study multiple peaks were detected in the plasma after oral and intravenous Sarawek et al. Table 4: Pharmacokinetic parameters of free luteolin after i.v. administration of 50 mg/kg luteolin. Data were fitted to a twocompartment model. Parameter Luteolin i.v. A (μg/mL) 9.6 B (μg/mL) 1.3 α (1/h) 1.9 β (1/h) 0.08 K12 (1/h) 1.2 K21 (1/h) 0.3 Ke (1/h) 0.48 Vc (L/kg) 4.5 Vt (L/kg) 18.2 Cl (L/h/kg) 2.2 t½α (h) 0.3 t½β (h) 9.1 t½ke (h) 1.4 AUC (h * µg/mL) 22.9 Cmax(µg/mL) 11.0 administration of luteolin, an enterohepatic recirculation of luteolin can be suggested; this is similar to other studies. Liu et al. [30] observed a rapid absorption and metabolism of aglycones such as apigenin and quercetin into phase II conjugates following enteric and enterohepatic recycling. Ma et al. [31] reported an enterohepatic recirculation of naringenin in rat plasma. However, our data did not show multiple peaks in the plasma concentration-time profile of free luteolin after intravenous and oral administration, presumably due to the limited number of data time points. In conclusion, after oral administration of luteolin, the compound was rapidly absorbed and metabolized in plasma. Moreover, plasma-concentration-time curves of luteolin metabolites revealed secondary peaks. The bioavailability of luteolin was low and the urinary excretion of luteolin and its conjugates did not dominate. Further studies are needed to confirm that the luteolin Cmax and AUC data reported here are genuinely associated with the threshold of its reported antitumorgenic, antiproliferative and xanthine oxidase inhibitory effects. In addition, Pharmacokinetic parameters of luteolin Natural Product Communications Vol. 3 (12) 2008 2033 further investigations are required to clarify if luteolin conjugates demonstrate pharmacological activity. at 5, 10, 15, 30, 45 min, and 1, 2, 4, 6, 12, and 24 h for oral administration. Prior to blood collection, the rats were anaesthetized with halothane and the blood loss was replaced with an equal volume of isotonic saline. The blood sample was centrifuged for 15 min at 4,000 rpm at 4oC. The supernatant, in aliquots of 0.20 mL, was transferred into tubes and 10 μL of 0.58 M acetic acid was added to each aliquot for stabilization. The plasma samples were stored at -80oC until analysis. Urine was collected over 24 h and an aliquot of 50 mL was mixed with 1 g ascorbic acid as antioxidant and stored at -80oC until analysis. Experimental Materials: Luteolin (99%) was purchased from Indofine Chemical Company, Inc. (Somerville, NJ, USA) and naringenin (≥ 96%) (internal standard) from Roth Carl Roth GmbH+Co. (Germany). Acetone, acetonitrile (CH3CN), acetic acid, dimethylsulfoxide (DMSO), methanol, and orthophosphoric acid (85% p.a.) were obtained from Fisher Scientific (Fair Lawn, NJ, USA). L (+)ascorbic acid (≥ 99.9%) was obtained from Acros Organics (New Jersey, USA). Trifluoroacetic acid was purchased from Fluka (Milwaukee, WI, USA) and β-glucuronidase/sulfatase (type HP-2, Helix pomatia), polyethylene glycol 200, and sodium dihydrogenphosphate monohydrate (NaH2PO4.H2O), from Sigma Chemical Company (St. Louis, MO, USA). All buffers and aqueous solutions were prepared with purified water obtained from a NANOPure® system from Barnstead (Dubuque, IA, USA). Animals: Male Sprague-Dawley rats, weighing 300-400 g were purchased from Harlan (IN, USA) and divided into the experimental groups, each containing 8-11 rats. The animals were housed in plastic cages and were allowed to adapt to their environment for one week before used for experiments. All the animals were maintained on a 12h/12h light/dark cycle. They received a standard chow and water ad libitum during the duration of the experiment. All animal experiments were performed according to the policies and guidelines of the Institutional Animal Care and Use Committee (IACUC) of the University of Florida, Gainesville, USA (NIH publication # 85-23). Methods: The pharmacokinetic studies were carried out by the sparse sampling approach wherein blood samples were collected from 8-11 rats. Luteolin was administered to two groups of rats (n = 8-11 in each group). Group one received a single i.v. dose of 50 mg/kg of luteolin via the tail vein. Group two received the same dose orally by gavage. Luteolin was dissolved in 30% DMSO and 70% polyethylenglycol (PEG) 200. Plasma samples (250 μL per blood sample) were collected from the sublingual vein into heparinized tubes at 3, 5, 10, 30 min, and 1, 2, 4, 6, 12 , and 24 h for i.v. injection, and Analytical methods: The plasma concentrations of unchanged free and conjugated luteolin in rat plasma and urine were determined by the method published earlier, with slight modifications [19]. Plasma and urine samples were analyzed using a Shimadzu VP series HPLC system (Kyoto, Japan) equipped with an SPD-M10Avp diode array detector. A Lichrospher® 100 RP-18 (5 μm. Merck KgaA) was used for the separation of luteolin. The column was kept at 25oC. The eluents were (A) 50 mM phosphate buffer (NaH2PO4, pH 2.1) and (B) CH3CN. The following solvent gradient was applied: 20% B (6 min) and 20-50% B (21 min).The gradient was followed by 10 min column flushing and post-run equilibration, respectively. Total run time was 40 min. The flow rate was 1 mL/min. Forty μL of each sample was injected into the RP-HPLC system. Chromatograms were acquired at 330 nm. For the determination of total luteolin, 10.0 μL internal standard (naringenin, 1.05 mg/mL), 10.0 μL 0.5% (m/v) ascorbic acid and 20.0 μL acetic acid (0.58 M) were added to 200.0 μL of the plasma sample, followed by the addition of 12.0 μL β-glucuronidase/sulfatase solution. The mixture was incubated at 37oC for 1 h. Protein was precipitated by adding 240 μL of acetone. The mixture was vortexed for 1 min and centrifuged for 15 min at 4000 rpm at 4oC. The supernatant was transferred to tubes containing 4.0 μL 0.5% (w/v) ascorbic acid and 8.0 μL 1 M trifluoroacetic acid, and evaporated to dryness in a vacuum centrifuge. The residue was reconstituted in 60 μL of methanol: water (1:1, v/v), centrifuged for 10 min at 13,200 rpm, and 40 μL was injected into the HPLC. For the determination of unchanged luteolin in plasma, the sample was extracted in the same manner as described above, without adding the enzyme. The concentrations of luteolin and its conjugates in urine samples were measured using the same method as described for the plasma, except urine samples were 2034 Natural Product Communications Vol. 3 (12) 2008 centrifuged for 15 min at 13,200 rpm after adding 240.0 μL acetone, and then 40.0 μL of the supernatant was injected into the HPLC. The conjugates (glucuronides or sulfates) of luteolin were calculated by subtracting total luteolin from unchanged luteolin. The calibration curve was linear in the examined concentration range of 0.1 to 50 μg/mL with (R2 ≥ 0.99), with the detection limit of 0.1 μg/mL. The inter-day and intra-day precisions were less than 15%. The extraction recoveries were more than 90% for all samples. Data analysis: Mean concentration of luteolin and its conjugates versus time curves were generated in Grapad Prim® (version 4.0, San Diego, CA). The pharmacokinetic parameters were determined by noncompartmental and compartmental analysis using WinNonlin software package, version 4.1, (Pharsight Corporation, USA). The mean data were used for the analysis. Non-compartmental PK: The PK parameters determined were the AUC, maximum concentration in plasma (Cmax), time to reach Cmax (Tmax), the elimination rate constant (ke), the elimination half life (t1/2), the volume of distribution (Vd) and the clearance (CL). AUC 0→last was calculated using a linear/log trapezoidal method from time zero to last sampling point equal to or above the lower limit of quantification. Both Cmax and Tmax were obtained from the plots of plasma concentration versus time. The ke was obtained by linear regression of the terminal log linear phase of the concentrationtime curve. The elimination half-life (t1/2) was determined as 0.693/ke. The volume of distribution area (Vdarea) was calculated using CL/ke. The clearance (Cl) was calculated as Dose/AUC and the systemic bioavailability (F %) was calculated as F % = (AUC p.o. /AUC i.v..) × 100. Compartmental PK: Luteolin concentrations showed a better fit in a two compartment body model compared to a one compartment body model. The equation for the two compartment model (Figure 2) is as followed: C = A.e-αt + B.e-βt (i.v.), where C is the concentration of drug in plasma at time t; A and B are the mathematical coefficient; α is the distribution rate Sarawek et al. constant; β is the elimination rate constant; and t is time. After i.v. administration, AUC 0-∞ was calculated using following equation: AUC 0-∞ i.v. = A/α + B/β. The elimination half life was calculated as ln (2/ke). The volume of distribution of central compartment (Vc) was calculated as Dose/Ke × AUC. The volume of distribution of peripheral compartment (Vt) was calculated as Vc × k12/k21. The clearance (Cl) was calculated as Dose/AUC. Goodness of fit was determined by the AIC (Akaike Criteria) and SC (Schwartz Criteria). The lower the AIC and SC, the more appropriate is the selected model. Validation: The method was validated over the range of concentration of luteolin present in plasma and urine. The validation parameters of linearity, sensitivity, specificity, precision, accuracy and stability were determined. Plasma: The calibration curve (n = 9) operating in the range of 100-10000 ng/mL for luteolin in rat plasma was linear (r2 > 0.99). The limit of quantification (LOQ) of luteolin in plasma was 100 ng/mL. The precisions intra- and inter-day for luteolin were satisfactory with CV values between 1.3 and 12.3%. Similarly, the accuracy of the assay obtained with quality control samples containing 300, 800, and 3000 ng/mL luteolin was between 94.2 and 106.3% of the nominal values. The mean recovery assessed at three distinct levels of concentration (100, 500 and 10000 ng/mL) ranged from 95.7 to 106.4% of the expected values. Urine: The calibration curve (n = 9) operating in the range of 500-50,000 ng/mL for luteolin in rat urine was linear (r2 > 0.99). The LOQ of luteolin in urine was 500 ng/mL. The precisions intra- and inter-day for luteolin were satisfactory with CV values between 0.30 and 13.25%. Similarly, the accuracy of the assay obtained with quality control samples containing 500, 3,000, and 10,000 ng/mL luteolin was between 98.21 and 109.28% of the nominal values. The mean recovery assessed at three distinct levels of concentration (500, 3,000 and 10,000 ng/mL) ranged from 99.56 to 112.23% of the expected values. References [1] Bravo L. (1998) Polyphenols: chemistry, dietary sources, metabolism, and nutritional significance. Nutrition Reviews, 56, 317-333. [2] Ross JA, Kasum CM. (2002) Dietary flavonoids: bioavailability, metabolic effects, and safety. Annual Review of Nutrition, 22, 19-34. [3] Ju W, Wang X, Shi H, Chen W, Belinsky SA, Lin Y. (2007) A critical role of luteolin-induced reactive oxygen species in blockage of tumor necrosis factor-activated nuclear factor-kappaB pathway and sensitization of apoptosis in lung cancer cells. Molecular Pharmacology, 71, 1381-1388. Pharmacokinetic parameters of luteolin Natural Product Communications Vol. 3 (12) 2008 2035 [4] Samy RP, Gopalakrishnakone P, Ignacimuthu S. (2006) Anti-tumor promoting potential of luteolin against 7,12dimethylbenz(a)anthracene-induced mammary tumors in rats. Chemico-Biological Interactions, 164, 1-14. [5] Coleta M, Campos MG, Cotrim MD, Lima TC, Cunha AP. (2008) Assessment of luteolin (3',4',5,7-tetrahydroxyflavone) neuropharmacological activity. Behavioral Brain Research, 189, 75-82. [6] Manju V, Balasubramaniyan V, Nalini N. (2005) Rat colonic lipid peroxidation and antioxidant status: the effects of dietary luteolin on 1,2-dimethylhydrazine challenge. Cellular & Molecular Biology Letters, 10, 535-551. [7] Manju V, Nalini N. (2005) Chemopreventive potential of luteolin during colon carcinogenesis induced by 1,2-dimethylhydrazine. The Italian Journal of Biochemistry, 54, 268-275. [8] Manju V, Nalini N. (2007) Protective role of luteolin in 1,2-dimethylhydrazine induced experimental colon carcinogenesis. Cell Biochemistry and Function, 25, 189-194. [9] Bagli E, Stefaniotou M, Morbidelli L, Ziche M, Psillas K, Murphy C, Fotsis T. (2004) Luteolin inhibits vascular endothelial growth factor-induced angiogenesis; inhibition of endothelial cell survival and proliferation by targeting phosphatidylinositol 3'-kinase activity. Cancer Research, 64, 7936-7946. [10] Kim JS, Lee HJ, Lee MH, Kim J, Jin C, Ryu JH. (2006) Luteolin inhibits LPS-stimulated inducible nitric oxide synthase expression in BV-2 microglial cells. Planta Medica, 72, 65-68. [11] Ferriola PC, Cody V, Middleton E. (1989) Protein kinase C inhibition by plant flavonoids. Kinetic mechanisms and structureactivity relationships. Biochemical Pharmacology, 38, 1617-1624. [12] Nagao A, Seki M, Kobayashi H. (1999) Inhibition of xanthine oxidase by flavonoids. Bioscience, Biotechnology and Biochemistry, 63, 1787-1790. [13] Sarawek S, Feistel B, Pischel I, Butterweck V. (2008) Flavonoids of Cynara scolymus possess potent xanthine oxidase inhibitory activity in vitro but are devoid of hypouricemic effects in rats after oral application. Planta Medica, 74, 221-227. [14] Boersma MG, van der Woude H, Bogaards J, Boeren S, Vervoort J, Cnubben NH, van Iersel ML, van Bladeren PJ, Rietjens IM. (2002) Regioselectivity of phase II metabolism of luteolin and quercetin by UDP-glucuronosyl transferases. Chemical Research in Toxicology, 15, 662-670. [15] Murota K, Shimizu S, Miyamoto S, Izumi T, Obata A, Kikuchi M, Terao J. (2002) Unique uptake and transport of isoflavone aglycones by human intestinal caco-2 cells: comparison of isoflavonoids and flavonoids. The Journal of Nutrition, 132, 1956-1961. [16] Chen T, Li LP, Lu XY, Jiang HD, Zeng S. (2007) Absorption and excretion of luteolin and apigenin in rats after oral administration of Chrysanthemum morifolium extract. Journal of Agricultural and Food Chemistry, 55, 273-277. [17] Li L, Jiang H, Wu H, Zeng S. (2005) Simultaneous determination of luteolin and apigenin in dog plasma by RP-HPLC. Journal of Pharmaceutical and Biomedical Analysis, 37, 615-620. [18] Shimoi K, Okada H, Furugori M, Goda T, Takase S, Suzuki M, Hara Y, Yamamoto H, Kinae N. (1998) Intestinal absorption of luteolin and luteolin 7-O-beta-glucoside in rats and humans. FEBS Letters, 438, 220-224. [19] Wittemer SM, Ploch M, Windeck T, Mueller SC, Drewelow B, Derendorf H, Veit M. (2005) Bioavailability and pharmacokinetics of caffeoylquinic acids and flavonoids after oral administration of Artichoke leaf extracts in humans. Phytomedicine, 12, 28-38. [20] Hollman PC, de Vries JH, van Leeuwen SD, Mengelers MJ, Katan MB. (1995) Absorption of dietary quercetin glycosides and quercetin in healthy ileostomy volunteers. American Journal of Clinical Nutrition, 62, 1276-1282. [21] Hollman PC, Katan MB. (1997) Absorption, metabolism and health effects of dietary flavonoids in man. Biomedicine and Pharmacotherapy, 51, 305-310. [22] Chen X, Yin OQ, Zuo Z, Chow MS. (2005) Pharmacokinetics and modeling of quercetin and metabolites. Pharmaceutical Research, 22, 892-901. [23] Romanova D, Grancai D, Jozova B, Bozek P, Vachalkova A. (2000) Determination of apigenin in rat plasma by high-performance liquid chromatography. Journal of Chromatography A, 870, 463-467. [24] Zhang L, Zuo Z, Lin G. (2007) Intestinal and hepatic glucuronidation of flavonoids. Molecular Pharmacology, 4, 833-845. [25] Wang Y, Cao J, Zeng S. (2005) Involvement of P-glycoprotein in regulating cellular levels of Ginkgo flavonols: quercetin, kaempferol, and isorhamnetin. Journal of Pharmacy and Pharmacology, 57, 751-758. [26] Cave NJ, Backus RC, Marks SL, Klasing KC. (2007) The bioavailability and disposition kinetics of genistein in cats. Journal of Veterinary Pharmacology and Therapeutics, 30, 327-335. [27] Nielsen SE, Young JF, Daneshvar B, Lauridsen ST, Knuthsen P, Sandstrom B, Dragsted LO. (1999) Effect of parsley (Petroselinum crispum) intake on urinary apigenin excretion, blood antioxidant enzymes and biomarkers for oxidative stress in human subjects. British Journal of Nutrition, 81, 447-455. [28] Gugler R, Leschik M, Dengler HJ. (1975) Disposition of quercetin in man after single oral and intravenous doses. European Journal of Clinical Pharmacology, 9, 229-234. [29] Walle T, Otake Y, Brubaker JA, Walle UK, Halushka PV. (2001) Disposition and metabolism of the flavonoid chrysin in normal volunteers, British Journal of Clinical Pharmacology, 51, 143-146. 2036 Natural Product Communications Vol. 3 (12) 2008 Sarawek et al. [30] Liu Y, Dai Y, Xun L, Hu M. (2003) Enteric disposition and recycling of flavonoids and ginkgo flavonoids. Journal of Alternative and Complementary Medicine, 9, 631-640. [31] Ma Y, Li P, Chen D, Fang T, Li H, Su W. (2006) LC/MS/MS quantitation assay for pharmacokinetics of naringenin and double peaks phenomenon in rats plasma. International Journal of Pharmaceutics, 307, 292-299. NPC Natural Product Communications Complete Characterization of Extracts of Onopordum illyricum L. (Asteraceae) by HPLC/PDA/ESIMS and NMR 2008 Vol. 3 No. 12 2037 - 2042 Luisella Verottaa*, Laura Belvisia, Vittorio Bertaccheb and Maria Cecilia Loic a Dipartimento di Chimica Organica e Industriale, Universita’ degli Studi di Milano, via Venezian 21, 20133 Milan, Italy b Istituto di Chimica Organica “A. Marchesini”, Universita’ degli Studi di Milano, via Venezian 21, 20133 Milan, Italy c Dipartimento di Scienze Botaniche, Università degli Studi di Cagliari, Viale Sant'Ignazio da Laconi, 13, 09123 Cagliari, Italy luisella.verotta@unimi.it Received: August 5th, 2008; Accepted: November 17th, 2008 The aerial parts of Onopordum illyricum L. (Asteraceae) are eaten raw in salad in the Mediterranean area, representing a food of good nutritional value. Extracts of different parts of this plant have been analyzed by HPLC/DAD/ESIMS and the major compounds identified by NMR spectroscopy. Fatty acids, sesquiterpene lactones, triterpenes and polyphenols (flavones and caffeoyl quinic acids) fully describe the plant metabolism during the vegetation year. All the metabolites are non toxic nutrients, and are reported in the literature to possess biological activities positive for health, confirming the beneficial use in the diet of this thistle Keywords: Onopordum illyricum, Asteraceae, HPLC/PDA/ESIMS analysis, polyphenols, caffeoylquinic acids. The development of dietary agents for chemoprevention is a highly attractive anticancer strategy, because of the long-standing exposure of humans to compounds of this type, their relative lack of toxicity, and the existence of encouraging epidemiological clues [1a,1b]. Polyphenols present in the diet, for example, are receiving increasing interest as chemopreventive agents because of the epidemiological association between nutrients rich in polyphenols and the prevention of diseases like cancer and stroke [1c]. Onopordum illyricum L., Asteraceae, (“cardo maggiore” or “onopordo maggiore”a type of thistle) is included in the daily diet of Sardinian people. In some villages in the Nuoro (Seulo, Torpè) and Campidano districts it is used as a vegetable (“kàrdu molentìnu” in campidanese idiom), young scapes and capitula being eaten raw in salad, as side dishes [2]. In folk medicine either a decoction or a tea of the whole plant is used as a digestive, cough sedative and in biliary diseases [2]. In Tempio Pausania village (Northern Sardinia) the decoction or the infusion of flowering tops is used for the alleged treatment of malarial fever as an antipyretic and for washing exanthematic skin [3]. Plants of the genus Onopordum are characterized by the presence of sesquiterpene lactones, flavonoids and lignans. Although a phytochemical study of this species was presented a few years ago [4a], we decided to investigate collections from different vegetative periods of the phenotype growing in Sardinia. O. illyricum is a wild plant spread in the Mediterranean region in Portugal and Albania [4b]. This study was carried out on two different collections, namely on the plant at full vegetation (June) and the plant dried in the field (July collection). Extracts of different parts (scapes and capitula) were obtained through multiple solvent extraction techniques (Table 1). Variations in extraction yields between the young plants and the old ones are plausible, due to the residual water content in materials dried in the 2038 Natural Product Communications Vol. 3 (12) 2008 Verotta et al. Table 1: Yields (g) of extractives from different parts of Onopordum illyricum, collected at different periods. Part (w) Scapes and leaves (419 g) Capitula ( 274 g) OI050603UT Scapes and leaves (473 g) OI310703UI Capitula (461 g) OI310703UI MeOH 14.0%* n-Hexane CHCl3 5.0 (1.2%)* 15.5 (3.7%) n-BuOH 11.5 (2.7%) 11.2% 4.2 (1.5%) 2.7 (1.0%) 7.3 (2.6%) 11.0% ** 14.9 (3.2%) 13.2 (2.7%) 7.2% ** 11.3 (2.4%) 9.5 (2.1%) * referred to the dry vegetable material ** total methanol extract was directly counter extracted with CHCl3 laboratory. A constant percentage is seen among the most polar extracts (n-BuOH), while the less polar ones show differences, mostly related to the presence of chlorophyll in young material. The n-hexane fraction contained fatty acids (mainly oleic and linoleic acids) [5] and triterpenes (taraxasteryl acetate and pseudotaraxasteryl acetate) as major components. The dichloromethane extract afforded onopordopicrin [6a,6b] as the most abundant constituent, accompanied by 8α-sarracinoyl salonitenolide as a by-product [6c]; these sesquiterpene lactones represented more than 70% of the total dichloromethane fraction. In this phenotype, onopordopicrin alone represents about 1% of the vegetable material. The lipophilic extracts of young scapes and capitula showed identical phytochemical profiles, which were also identical to those of the aged material, with a clear difference between the yield of fatty acids and sterols and the sesquiterpene lactones that are more abundant in the young vegetable material. The n-BuOH extract from young scapes was separated into ten fractions by chromatography on Sephadex LH20. They were further purified by preparative reversed phase HPLC to give two dicaffeoylquinic acids [M+ 516 m/z] and a succinyl dicaffeoylquinic [M+ 616 m/z] derivative. Comparison of the spectroscopic data of the isolated compounds with those reported in the literature allowed the assignment of the structures as 3,5-di-Ocaffeoylquinic acid (1) [7a,7b], and 1,5-di-Ocaffeoylquinic acid (3) [7c]. The structure of the triester (2) was tentatively assigned as 1-succinyl 3,5dicaffeoylquinic acid by 1D- and 2D- NMR experiments. The quinic acid ring was shown to be esterified at positions 3 and 5 (proton chemical shifts higher than 5.00 ppm), while the proton at position 4 remained unchanged with respect to the original value, for example in 5-caffeoyl quinic acid Compound (2) Table 2: 1H NMR (400 and 500 MHz) assignments δ, m, J (Hz) for compounds 2 and 3. Proton 8” (2) Acetone-d6 7.67 d, 15.9 7.60 d, 15.9 7.24 d, 2.0 7.22 d, 2.0 7.10 dd, 8.0, 2.0 7.05 dd, 8.0, 2.0 6.89 dd, 8.0, 3.1 6.88 dd, 8.0, 3.1 6.40 d, 15.9 8’ 5 6.30 d, 15.9 5.52 m 6.28 d, 15.9 6.37, d, 6.21 d, 15.9 15.9 6.23 d, 15.9 6.33 d, 15.9 6.21 d, 15.9 5.25 m 5.47 m 5.24 bt, 8.3 3 5.46 dt, 5.27 m 4 3.63 bt, 8.0 6eq 4.04 dd, 8.6, 3.85 dd, 8.5, 3.96 dd, 3.5 3.3 8.5, 3.3 2.60 m 2.46 m 2.60 m 2eq 2.60 m 2.43 m 2.77 m 2.28 m 6ax 2.01 m 2.01bt, 12.1 1.94 bt, 12.0 2ax 2.60 m 1.94 dd, 10.6, 2.0 2.43 m 2’’’ 3’’’ 2.60 m 2.60 m 2.43 m 2.43 m 2.52 m 2.52 m 7” 7’ 2” 2’ 6” 6’ 5” 5’ (2) DMSO-d6 7.50 d, 15.9 7.50 d, 15.9 7.07 bs 7.07 bs 7.01 d, 8.0 7.01 d, 8.0 6.78 d, 8.0 6.78 d, 8.0 (2) CD3OD 7.65 d, 15.9 7.63 d, 15.9 7.10 d, 2.0 7.08 d, 2.0 7.01dd, 8.0, 2.0 7.01 dd, 8.0, 2.0 6.82, d, 8.0 (3) DMSO-d6 7.48, 15.9 7.48 d, 15.9 7.03 d, 2.0 7.03 d, 2.0 6.97 dd, 8.0, 2.0 6.97 dd, 8.0, 2.0 6.78 d, 8.2, 2.0 6.80 d, 8.0 6.78 d, 8.2 (3) CD3OD 7.60 d, 15.9 7.60 d, 15.9 7.07 d, 2.0 7.07 d, 2.0 6.98 dd, 8.0, 2.0 6.98 dd, 8.0, 2.0 6.80 d, 8.0 6.32 d, 15.9 5.47 m 2.48 m 4.09 bt, 8.0 2.34 m 2.28 m 6.80 d, 8.0 6.28 d, 15.9 5.41 dt, 8.3, 3.5 4.31 ddd, 8.5, 8.0, 3.7 3.79 dd, 7.9, 3.3 2.59 dd, 13.2, 2.9 2.51, dd, 15.2, 3.4 2.08 dd, 13.2, 8.7 2.44 dd, 15.2, 4.4 (chlorogenic acid), where it resonates below 4.0 ppm. The diagnostic resonance of carbon 1 (around 80 ppm) indicated esterification of the hydroxyl group present on this carbon atom. Several attempts to determine either heteronuclear multiple bond correlations from the caffeoyl or succinyl side chain carbons to ring protons, or vice versa were made, even recording spectra in different solvents (see Tables 2 and 3), in order to ameliorate chemical shifts differentiation and solvent effects. However no information was obtained on the correct ring substitution, even by reproducing the experimental conditions reported for 3,5 dicaffeoylquinic acid [7a]. HPLC/DAD/ESIMS analysis of extracts of Onopordum illyricum Table 3: 13C NMR (100 and 125 MHz) assignments (δ, m) for compounds 2 and 3. Carbon 1 2 3 4 5 6 7 1’ 1’’ 2’ 2’’ 3’ 3’’ 4’ 4’’ 5’ 5’’ 6’ 6’’ 7’ 7’’ 8’ 8’’ 9’ 9’’ 1’’’ 2’’’ 3’’’ 4’’’ (2) DMSO-d6 80.0 s 32.3 t 71.8 d 70.0 d 70.3 d o* 173.7 s 125.9 s 125.9 s 115.4 d 115.4 d 146.1s 146.1 s 149.0 d 149.0 d 116.3 d 116.3 d 121.8 d 121.7 d 145.7 d 145.7 d 114.6 d 114.6 d 166.5 s 165.6 s 172.1 s 29.1 t 29.4 t 172.2 s (2) Acetone-d6 78.9 32.2 71.2 69.9 69.9 36.1 173.5 126.7 126.7 114.4 114.5 146.3 145.6 148.5 148.5 115.5 115.5 121.9 121.7 146.0 145.4 114.6 114.5 165.5 166.1 171.4 29.5 29.5 171.4 (3) DMSO-d6 79.8 34.6 67.9 71.4 70.5 36.2 173.0 126.0 126.0 114.8 114.8 145.8 145.7 148.9 148.9 115.3 115.3 121.8 121.8 146.0d 146.0d 114.7 114.7 166.4 165.7 o* =overlapped Figure 2: Lowest minimum energy conformation for compound 2. The arrow indicates the hydrogen bonding. Superimposable chemical shifts and shape of the proton spectra of the aromatic and quinic acid region of the triester with those of 3,5-di-O-caffeoylquinic acid (1) isolated in this work and by comparison with literature data [7a], tentatively assigned compound 2 as 1-succinyl 3,5-di-O-caffeoylquinic acid. Partial alkaline hydrolysis of 2 [7d] afforded a complex mixture of compounds where 3,5-di-O-caffeoylquinic acid was identified by TLC. It becomes apparent from an unconstrained Monte Carlo/Energy Minimisation (MC/EM) conformational search [7e] by molecular mechanics methods that the lowest minimum energy conformation (the 1C4 chair) Natural Product Communications Vol. 3 (12) 2008 2039 is that originally reported for known quinic acid diand tri-esters. It accommodates the succinyl moiety in an axial position, the conformational asset being further supported by the existence of hydrogen bonding between the succinyl terminal carboxy group and the 5-O-caffeoyl carbonyl moiety (Figure 2). To characterize the complete compositions of the n-butanol extracts of scapes and capitula collected at different vegetative periods, they were analyzed by HPLC/PDA/ESIMS. Accordingly, flavonoid glycosides are eluted first, followed by the group of caffeoylquinic acids and the flavonoids at the end [8]. As can be seen in Figures 1A-D and Table 4, caffeoylquinic acids are the dominant phenolic compounds accumulating in the young material. The presence of important nutritional biomarkers like fatty acids, sesquiterpene lactones, triterpenes, flavones and their glycosides and caffeoylquinic acid derivatives justifies the use of O. illirycum as a beneficial food with chemopreventive potential. All the identified compounds in fact show interesting in vitro and in vivo bioactivities. Onodorpicrin, a sesquiterpene lactone, present in the leaves of Arctium lappa L. [6b], has an IC50 value of approximately 15 µM in cellular lineage of promyelocytic leukemia (HL60) used as a model for antitumor studies. In classic models of ulcer induction the compound also presented significant antiulcerogenic activity. The data also reveal that pretreatment with onopordopicrin is able to reduce intestinal inflammation in a model of colitis in rats, suggesting an excellent potential for therapy in the gastrointestinal area [9]. Taraxasterol inhibits tumor promotion, invasion of tumor cells and metastasis. The acetate has weaker activity [10,10b]. Mono- and di-caffeoylquinic acids play a key-role in the overall anti-oxidant/health value of globe artichoke [10c]. Caffeoylquinic acids have been implicated in the inhibition of HIV integrase, a key player in HIV replication and its insertion into host DNA [10d], the protection of proteins, lipids and DNA from oxidative damage caused by free radicals [10e], hepatoprotective, choleretic, diuretic, bile-expelling, and antibacterial and antifungal activities [11a,11b]. A full description of the phytochemical profile of O. illirycum has been reported. On this basis we can assert that the presence of this plant in the daily diet can be considered generally beneficial to human health, thus confirming that people living in the Mediterranean area still maintain a strong knowledge of the traditional uses of plants. 2040 Natural Product Communications Vol. 3 (12) 2008 Verotta et al. Table 4: HPLC/PDA/ESIMS of the n-BuOH extract of Onopordum illyricum aerial parts. Compound RT (min) HPLC-DAD λmax (nm) [M-H]-, [2M-H]- m/z Molecular formula Identification 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 11.75 19.93 20.30 20.90 22.36 22.88 24.25 25.11 26.35 28.22 29.29 31.65 32.78 37.65 38.77 219, 328 245, 335 245, 355 250, 350 245, 325 220,240, 325 240, 325 240, 330 245, 325 245, 325 235, 285 255, 270, 345 255, 265, 345 235, 275, 335 235, 265, 335 353 577 623 461 515 515 515 615 615 515 287, 575 315, 630 285, 571 299, 598 269, 539 C16H18O9 C27H30O14 C28H32O16 C22H22O11 C25H24O12 C25H24O12 C25H24O12 C29H30O15 C29H30O15 C25H24O12 C15H12O6 C16H12O7 C15H10O6 C16H12O6 C15H10O5 Chlorogenic acid Apigenin rutinoside Rhamnetin rutinoside Kaempferide glucoside 1,5-Dicaffeoylquinic acid 3,5-Dicaffeoylquinic acid Dicaffeoylquinic acid 1-Succinyl, 3,5-dicaffeoylquinic acid Succinyldicaffeoylquinic acid Dicaffeoylquinic acid Dihydrokaempferol Rhamnetin Luteolin 4’-OMe apigenin Apigenin 6 A 8 B 7 9 C 5 1 3+4 2 10 11 14 12 13 D 15 Figure 1: HPLC/PDA/ESIMS analyses of Onopordum illyricum n-butanol extracts (λ = 350 nm). Scapes (A), capitula (B), dry scapes (C), dry capitula (D). Experimental Plant material: This was collected at Uta (Cagliari district) in June (fresh material) and July (field dried material) 2003, and identified by M.C. Loi, Department of Botanical Science, University of Cagliari, Italy, where a voucher specimen was deposited. The fresh materials were dried at room temperature. Analytical and prep. HPLC: A) Waters 515 HPLC pump, equipped with a Waters 2487 Dual λ Absorbance Detector (fixed wavelengths 280, 330 nm) and a Waters Symmetry Shield RP18 5μm (4.6 x 250) column, eluted with a gradient of 0.1% CF3COOH in H2O (solvent A) and 0.1% CF3COOH/MeOH (solvent B), starting from 55% solvent A for 16 min, to 100% solvent B, at a flow rate 1 mL/min. 20 μL of solutions containing 5 mg/mL were injected. On the same instrument, the preparative separations were obtained through a Waters Symmetry Prep RP18 7 μm (190 mm x 150 mm), at a flow rate of 4 mL/min, eluted under isocratic conditions (25 min) with H2O-MeOH 70:30 and 0.1% di CF3COOH. B) Alternatively, a gradient was used of 0.1% CF3COOH in H2O (solvent A) and 0.1% CF3COOH/CH3CN (solvent B), starting from 90% solvent A to 90% solvent B in 60 min, at a flow rate of 1 mL/min. HPLC-PDA-MS analyses: A Thermo Finnigan instrument LCQ Advantage Series (Thermo Finnigan, San Jose, CA, USA), equipped with a quaternary pump (Surveyor), a diode-array detector, an electrospray ionisation source (ESI) and an ion HPLC/DAD/ESIMS analysis of extracts of Onopordum illyricum Natural Product Communications Vol. 3 (12) 2008 2041 trap analyser (negative mode) was used for LC–MS analyses and to obtain UV–vis and mass spectra of eluted compounds. The n-BuOH extracts and the single fractions from its purification (see below) were carried out using a Symmetry Shield (Waters) RP 18 250 x 4.6 mm I.D. (5 μm) column maintained at 30 °C by a column block heater (model 7970, Hichrom Ltd., Reading, UK). A linear gradient from 90:10 to 30:70 v/v (H2O:CH3CN) with 0.1% w/v TFA and a flow rate of 1mL/min was used. Samples were dissolved in the mobile phase and injected through a Rheodyne (model 7125) valve equipped with a 20 μL loop. Spectral data for the wavelength range 190 to 600 nm, using a photodiode-array detector (Surveyor), and integrated areas under the peaks detected at 350 nm were acquired. Three dimensional chromatograms using Thermo Xcalibur software (Thermo Finnigan) were recorded. pseudotaraxasteryl acetate and taraxasteryl acetate (84 mg). Fractions eluted with EtOAc (290 mg) were further purified on silica gel, eluted with CHCl3/i-PrOH 9:1 (400 mL), giving a mixture of oleic and linoleic acids (190 mg). Taraxasteryl acetate and pseudotaraxasteryl acetate were identified in a mixture by comparison of their IR, 1H, 13C NMR spectra with literature data. [11c,12]. Extraction and purification: Scapes and leaves (OI050603UT, 419 g) were reduced to powder with a robot mixer and extracted through percolation with MeOH at room temperature (4 x 1.8 L). After solvent evaporation, 58.5 g (14%) of total extract was obtained, which was added to MeOH (60 mL) and water (300 mL), filtered to remove a sticky material, and extracted with light petroleum (2 x 213 mL), then CH2Cl2 (2 x 213 mL), and n-butanol (4 x 300 mL). The organic phases were dried (Na2SO4) and evaporated to dryness under vacuum. Yields are reported in Table 1. Extractions were controlled through TLC, developed with CHCl3-MeOH (9:1), and revealed through either UV absorption or by spraying with methanolic H2SO4 10%. The n-butanol fraction was monitored by TLC using the organic phase of the solvent system CHCl3MeOH-n-PrOH-H2O 5:6:1:4 and revealed through either UV absorption or by spraying with FeCl3. The same procedure was followed to extract capitula (OI050603UT 274 g). Yields are reported in Table 1. The dried vegetable material collected (OI310703UI) followed a slightly different extraction procedure avoiding the counter extraction with light petroleum, mainly because of the absence of chlorophyll. Yields are reported in Table 1. The light petroleum extract (2 g) was purified by column chromatography on silica gel (100 g), eluted with CH2Cl2 (780 mL), CH2Cl2-EtOAC 95:5 (810 mL) and EtOAc (270 mL). Fractions were pooled according to their composition. One of the fractions eluted with CH2Cl2 (230 mg) was repeatedly crystallized from EtOAc, giving a mixture (1:3) of The dichloromethane extract (5.2 g) was purified by column chromatography on silica gel (120 g) eluted with CH3Cl-MeOH 95:5 (720 mL). Fractions 7-15 (3.2 g) were further purified on silica gel (flash column) eluted with light petroleum-EtOAc (2:3), yielding 2.4 g onopordopicrin (1) [Rf= 0.22, EtOAclight petroleum (70:30) (42.5%)] and 0.32 g 8αsarracinoyl salonitenolide [Rf= 0.21, EtOAc-light petroleum (70:30)]. The n-butanol extract (5 g) was purified by chromatography on Sephadex (2.8 x 100 cm) eluted with MeOH (1 L), at a flow rate of 15 mL/min. Fractions were pooled according to their profile on TLC (CHCl3-MeOH-n-PrOH-H2O 5:6:1:4) and revealing through either UV absorption or by spraying with FeCl3): 1-57 (3.13 g), 58-74 (0.323 g), 75-80 ( 0.057 g), 81-99 (0.426 g), 100-105 (0.197 g), 106-119 (0.183 g), 120-136 (0.295 g), 140-143 (0.05 g), 144-167 (0.1192 g), 167-184 (0.043 g) Fraction 81-99 was submitted to prep HPLC according to method A. 10 mg each of two compounds (1) (Rt 20.75 min, method A; 23.25, method B) identified as 3,5 dicaffeoylquinic acid [7a,7b], and (2) ([α]25D = - 30.1 (c 0.5, MeOH); UV λmax (nm) 240, 330) (Rt 26.17 min, method A; 25.20, method B) were isolated. Fraction 106-119 was submitted to prep HPLC according to method B. 22 mg of 1,5-di-O-caffeoylquinic acid (3) [7c] (Rt 22.85, method B) was isolated. Computational studies: Conformational preferences of 1-succinyl 3,5-di-O-caffeoylquinic acid (compound 2) were investigated by molecular mechanics calculations within the framework of MacroModel version 9.1 (Macromodel), using the MacroModel implementation of the MM2 force field [13] (denoted MM2*) and the implicit water GB/SA solvation model [14a]. The torsional space of the compound was randomly varied with the usagedirected Monte Carlo conformational search of Chang, Guida, and Still [7e]; 1000 starting structures for each variable torsion angle were generated and minimized until the gradient was less than 0.05 2042 Natural Product Communications Vol. 3 (12) 2008 kJ/Åmol, using the truncated Newton-Raphson method [14b] implemented in MacroModel. Duplicate conformations, and those with energy greater than 6 kcal/mol above the global minimum were discarded. Verotta et al. Acknowledgments – Mrs Manuela Gilberti is gratefully acknowledged for technical assistance in recording NMR experiments. References [1] (a) Khan N, Afaq F, Mukhtar H. (2008) Cancer chemoprevention through dietary antioxidants: progress and promise. Antioxidants & Redox Signaling, 10, 475-510.; (b) Aggarwal BB, Shishodia S. (2006) Molecular targets of dietary agents for prevention and therapy of cancer. Biochemical Pharmacology, 71, 1397-1421; (c) Han X, Shen T, Lou H. (2007) Dietary polyphenols and their biological significance. International Journal of Molecular Sciences, 8, 950-988. [2] Atzei AD. (2003) Le piante nella tradizione popolare della Sardegna. Carlo Delfino Ed. [3] Ballero M, Bruni A, Sacchetti G, Poli F, (1997) - Le piante utilizzate nella medicina popolare nel Comune di Tempio (Sardegna settentrionale). Acta Phytotherapeutica, 1, 23-29. [4] (a) Braca A, De Tommasi N, Morelli I, Pizza C. (1999) New metabolites from Onopordum illyricum. Journal of Natural Products, 62, 1371-1375; (b) Tutin TG, Heywood VH, Burges NA, Moore DM, Valentine DH, Walters SM, Webb DA. (Eds) (1976) Flora Europaea, 4, 247, Cambridge University Press. [5] Aitzetmüller K, Ivanov SA. (1998) Fatty http://sofa.bfel.de/cgi/sofa/lds.pl?TAB_ID=TYP2_00005 [6] (a) Lonergan G, Routsi E, Georgiadis T, Agelis G, Hondrelis J, Matsoukas J, Larsen LK, Caplan FR. (1992) Isolation, NMR studies, and biological activities of onopordopicrin from Centaurea sonchifolia. Journal of Natural Products, 55, 225-228; (b) Savina AA, Sheichenco VI, Stikhin YV, Stichin VA, Sokol’skaya TA, Anisimova OS, Kopyt’ko YA, Grodniskaya EI, Cherkasov OA (2006) Sesquiterpene lactones in juice of great burdock leaves. Pharmaceutical Chemistry Journal, 40, 624-626; (c) El-Marsy S, Darwish FA, Abou-Dona A, Abou-Karam MA, Grenz M. (1985) Sesquiterpene lactones from Centaurea glomerata. Phytochemistry, 24, 999-1001. [7] (a) Pauli GF, Poetsch F, Narhstedt A . (1998) Structure assignment of natural quinic acid derivatives using proton nuclear magnetic resonance techniques. Phytochemical Analysis, 9, 177-185; (b) Wu C, Chen F, Wang X, Wu Y, Dong M, He G, Galyean RD, He L, Huang G. (2007) Identification of antioxidant phenolic compounds in feverfew (Tanacetum parthenium) by HPLC-ESI-MS/MS and NMR. Phytochemical Analysis, 18, 401-410; (c) Merfort I. (1992) Caffeoylquinic acids from flowers of Arnica montana and Arnica chamissonis. Phytochemistry, 31, 2111-2113; (d) Leung J, Fenton TW, Clandinin DR. (1981) Phenolic components of sunflower flour. Journal of Food Science, 46, 1386-1389; (e) Chang G, Guida WC, Still WC. (1989) An internal-coordinate Monte Carlo method for searching conformational space. Journal of the American Chemical Society, 111, 4379-4386. [8] Lai J-P, Lim YH, Su J, Shen H-M, Ong CN. (2007) Identification and characterization of major flavonoids and caffeoylquinic acids in three Compositae plants by LC/DAD-APCI/MS. Journal of Chromatography B, 848, 215-225. [9] Albino de Almeida AB. (2005) Atividade antiulcerogenica e antiinflamatoria intestinal da Arctium lappa. PhD thesis, Universidade Estadual de Campinas. Campinas, SP. Brasil. [10] (a) Ovesna Z, Vachalkova A, Horvathova K. (2004) Taraxasterol and β-sitosterol: new naturally occurring compounds with chemoprotective/ chemopreventive effects: minireview. Neoplasma, 51, 407-414.; (b) Takasaki M, Konoshima T, Tokuda H, Masuda K, Arai Y, Shiojima K, Ageta H.(1999) Anti-carcinogenic activity of Taraxacum plant. II. Biological & Pharmaceutical Bullettin, 22, 606-610; (c) Wang MF, Simon JE, Aviles IF, He K, Zheng QY, Tadmor Y. (2003) Analysis of antioxidative phenolic compounds in artichoke (Cynara scolymus L.). Journal of Agricultural and Food Chemistry, 51, 601-608; (d) Slanina J, Taborska E, Bochorakova H, Slaninova I, Humpa O, Robinson WE, Schram H. (2001) New and facile method of preparation of the antiHIV-1 agent, 1,3-dicaffeoylquinic acid. Tetrahedron Letters, 42, 3383-3385; (e) Perez-Garcia F, Adzet T, Canigueral S. (2000) Activity of artichoke leaf extract on reactive oxygen species in human leukocytes. Free Radical Research, 33, 661-665. [11] (a) Dogan S, Turan Y, Erturk H, Arslan O. (2005) Characterization and purification of polyphenol oxidase from artichoke (Cynara scolymus L.). Journal of Agricultural and Food Chemistry, 53, 776-785; (b) Zhu XF, Zhang HX, Lo R. Phenolic compounds of artichoke (Cynara scolymus L) and their antimicrobial activities. (2004) Journal of Agricultural and Food Chemistry, 52, 7272-7278; (c) Reynolds WF, McLean S, Poplawski J, Enriquez RG, Escobar LI, Leon I. (1986) I. Total assignment of 13C and 1H spectra of three isomeric triterpenol derivatives by 2D NMR: an investigation of the potential utility of 1H chemical shifts in structural investigations of complex natural products. Tetrahedron, 42, 3419-3428. acids from Onopordum illyricum. Unpublished results [12] Matsunaga S, Tanaka R, Akagi M. (1988) Triterpenoids from Euphorbia maculata, Phytochemistry, 27, 535-537. [13] Allinger NL. (1977) Conformational analysis. 130. MM2. A hydrocarbon force field utilizing V1 and V2 torsional terms. Journal of the American Chemical Society, 99, 8127-8134. [14] (a) Still WC, Tempczyk A, Hawley RC, Hendrickson T. (1990) Semianalytical treatment of solvation for molecular mechanics and dynamics. Journal of the American Chemical Society, 112, 6127-6129; (b) Ponder JW, Richards FM. (1987) An efficient Newtonlike method for molecular mechanics energy minimization of large molecules. Journal of Computational Chemistry, 8, 1016-1024. NPC Natural Product Communications Phenolic Profiles of Four Processed Tropical Green Leafy Vegetables Commonly Used as Food 2008 Vol. 3 No. 12 2043 - 2048 Sule Ola Salawua, Marzia Innocentib, Catia Giaccherinib , Afolabi Akintunde Akindahunsia and Nadia Mulinacci*,b a Department of Biochemistry, Federal University of Technology, P.M.B. 704, Akure, Nigeria b Department of Pharmaceutical Science, and CeRA University of Florence, Sesto F.no, (FI), Italy nadia.mulinacci@unifi.it Received: July 23rd, 2008; Accepted: October 28th, 2008 The phenolic profiles are presented of four tropical green leafy vegetables (Ocimum gratissimum, Vernonia amygdalina, Corchorus olitorius and Manihot utilissima) commonly used as food, after application of traditional treatments, such as boiling and abrasion. The HPLC/DAD/MS technique was mainly used to carry out this study. Preliminary evaluation of the antioxidant properties of the vegetables was also performed using the DPPH in vitro test. For the first time, seasonal variations in the phenolic content of the four investigated vegetables were highlighted. Of the four plants, all showed only quantitative differences, except for Ocimum graticimum, in which cichoric acid, previously detected as one of the main constituents of this vegetable collected in November (dry season), was absent in the sample harvested in March. The phenolic constituents are chemically unmodified after a strong heating process, such as the traditional blanching (about 15 minutes) applied by Nigerian people prior to consuming these vegetables. Nevertheless, these typical preparations showed a consistent decrease in the total phenolic compounds with respect to the raw material, particularly for Corchorus olitorius (from 42.3 to 5.56 mg/g dried leaves) and Vernonia amygdalina (from 40.2 to 4.4 mg/g dried leaves). As expected, when the blanching treatment is reduced to a few minutes, as for Manihot utilissima leaves, the cooked vegetable maintained almost unaltered its original phenolic content (around 10 mg/g dried leaves). The unique exception is the blanched Ocimum gratissimum sample that showed a consistent increment of the total phenols, particularly of rosmarinic acid (from 6.1 to 29.8 mg/g dried leaves) with respect to the unprocessed vegetable. Keywords: Ocimum gratissimum, Vernonia amygdalina, Corchorus olitorius, Manihot utilissima, phenolic compounds, traditional preparations, HPLC/DAD/MS. Most of the compositional aspects of vegetables commonly used in the Western diet are well known, but scant data are available on endemic plants from African regions. The present study attempts to improve knowledge of the phenolic content of the processed leaves of four vegetables commonly used for food and medicinal purposes in Nigeria. Ocimum gratissimum L. (Og) (Labiateae), Manihot utilissima Pohl. (Mu) (Euphorbiaceae), Vernonia amygdalina L. (Va) (Compositae), and Corchorus olitorius L. (Co) (Tiliaceae) are considered in the present work. Among them, Og, Va and Co are mainly consumed as fresh or pot vegetables. The tuber of Mu is the part mainly eaten, but the young leaves are gaining acceptance also as a pot vegetable. Ocimum gratissimum, African basil, and known as “efinrin” by Nigerian people, is usually collected from May to October. With regards to its composition, the principal data are related to its flavonoid composition [1], but mainly related to plants grown in the UK [2]. The authors found that the profile of the main flavonoids was similar in all accessions belonging to the same species and they showed vicenin-2, luteolin 7-O-glucoside, quercetin 3-O-glucoside and quercetin 3-O-rutinoside as constituents of the leaves, together with xanthomicrol, cirsimaritin, and kaempferol-3-Orutinoside. For medicinal properties, in south-western Nigeria, Og is mainly known for its antimicrobial activities against bacteria causing diarrhea [3]. 2044 Natural Product Communications Vol. 3 (12) 2008 Manihot utilissima or cassava, known by the local population as “ewe ege”, is harvested throughout the year. This vegetable is widespread in Nigeria, which is the world's largest producer. Though less popular in the Nigerian diet compared with other vegetables, the dietary acceptance of Mu leaves has been increasing within local populations [4]. Vernonia amigdalina leaves, known as “ewuro” by the local population, are harvested throughout the year and are characterized by an intense bitterness. Previous reports on the composition of this plant highlighted the presence of luteolin, luteolin 7-O-βglucoside and luteolin 7-O-β-glucuronide as main flavonoids, together with several dicaffeoyl derivatives [5]. Also, some saponins and sesquiterpene lactones have been reported in the leaves [6]. Due to its documented antimalarial [7-9], antimicrobial [10-11] and anticancer properties [12], Va is probably one of the most used medicinal plants in Nigeria. Corchorus olitorius or “Tossa Jute”, is quite popular for its leaves, which are usually collected from May to December and used as a pot herb. Jute leaves, also known as Jew's Mallow, are popular in West Africa and the Yoruba people of Nigeria call it "ewedu". The leaves are made into a common mucilaginous soup or sauce in some West African cooking traditions. C. olitorius leaves contain kaempferol glycosides, rutin and isoquercitrin [13], together with chlorogenic acid and several dicaffeoyl derivatives of quinic acid [5]. With regard to its use as a medicinal plant, this vegetable is mainly known in Nigeria for its laxative activity and as a blood purifier [14]. The present study was mainly focused on comparing the phenolic profile of the four vegetables obtained in different seasons to determine the qualitative and quantitative content of these compounds in the processed vegetables after application of traditional treatments, and to evaluate some antioxidant properties by using the DPPH in vitro test. Phenolic distribution in the unprocessed leaves. To recover all the main phenolic constituents, the dried leaves were extracted with ethanol/acidic water (7:3) at room temperature, and the extracts were then analyzed using a previously optimized chromatographic method [5]. The identification of the components of the extract was carried out by comparison with our previous results [5], with the help of their retention times, and UV-Vis and MS Salawu Sule et al. spectra. When necessary, the use of standard reference compounds and/or laboratory extracts helped to complete this identification. Our previous findings, which highlighted for the first time the phenolic composition of these vegetables, took into account samples collected in Nigeria during the dry season (November), while in this study we analyzed plants collected in March. Given that these vegetables are consumed throughout the year by the local population, it is of interest to evaluate if seasonality affects the phenolic composition of the vegetables. Regarding Co, its chromatographic profile at 330 nm is similar to that obtained for the sample collected in November, even if an increment of the ratio between the more polar monocaffeoyl quinic derivative (Co1) and the 1,5 dicaffeoyl quinic acid (Co 5) is observed. It is reasonable to hypothesize that these two molecules are biosynthetically correlated and that different climatic conditions can modulate their production in the plant. The dominant compound detected in the Va sample was 1,5 dicaffeoyl quinic acid (Va 10), differently from the findings observed for the leaves collected in November. At the same time, almost all the flavonoids, both luteolin and apigenin glycosides, were minor constituents. Among the four plants, the chemical profile of Og is the most complex due to the co-presence of metabolites belonging to different chemical classes (Table 1d). Major differences were observed between the leaves collected in the two seasons. In the March sample, together with a consistent amount of rosmarinic acid and the presence of the polar glycoside, vicenin 2 or apigenin 6,8-di-C-glucoside, a high concentration of the metoxyflavone, nevadensin, has again been highlighted [5]. Nevertheless, it is worth noting that cichoric acid, one of the main phenolic constituents previously detected and estimated at nearly 2.5 mg/g in the dried leaves collected in November [5], was completely absent in this sample. In the light of this finding, it can be said that, differently from the other phenolic constituents, the biosynthesis of this dicaffeoyl tartaric acid in African basil is particularly sensitive to seasonal variation. Contrary to the HPLC profiles observed at 330 nm for Va and Og, that for Mu seemed to be unaffected by the time of harvest. In fact, the relative ratios between the four main Phenolic profiles of four tropical green leafy vegetables Natural Product Communications Vol. 3 (12) 2008 2045 Table 1a Compounds of Corcorus olitorius Raw Blanched Co1 - caffeoyl quinic derivative 14.8±0.4 2.8±0.03 Co2 - chlorogenic acid 0.2±0.02 0.07±0.00 Co3+4 - hyperoside+isoquercitrin 2.1±0.06 0.3±0.004 Co5 1, 5 -dicaffeoyl quinic acid 23.2±0.6 2.0±0.03 Co6 - dicaffeoyl quinic acid 0.1±0.004 0.002±0.002 Co7 - dicaffeoyl derivative 1.7±0.03 0.5±0.004 Co8 - quercetin derivative 0.1±0.009 nd Table 1b Compounds of Vernonia amigdalina Raw Blanched Va1+2 - caffeoyl quinic and chlorogenic acids 2.3±0.3 0.4±0.02 Abrasion 0.5±0.04 Va4 - rutin 0.2±0.04 nd nd Va5 - luteolin 7-O-glu 2.7±0.3 0.3±0.02 0.4±0.02 Va7 - flavonoid 0.3±0.03 0.1±0.009 0.005±0.006 Va9 - luteolin 7-O-glucuronide 4.9±0.5 0.7±0.02 0.7±0.04 Va10 -1,5 dicaffeoyl quinic acid 23.47±3.0 1.4±0.2 6.9±1.5 Va11 - dicaffeoyl quinic acid 2.6±0.1 0.3±0.02 0.5±0.08 Va12 - dicaffeoyl quinic acid 2.5±0.4 0.4±0.03 1.3±0.5 Va13 - apigenin-O-glucuronide 0.7±0.02 0.4±0.02 0.2±0.02 Va14 - luteolin 0.2±0.03 0.1±0.01 0.6±0.06 Va15 - flavonoid 0.1±0.02 0.1±0.01 0.02±0.004 Va16 - flavonoid 0.09±0.01 0.04±0.004 0.3±0.01 Va17 - flavonoid 0.06±0.01 0.1±0.001 0.0015±0.0 Table 1c Compounds of Manihot utilissima Raw Mu1 - rutin 5.1±0.2 Blanched 5.8±1.0 Mu2 - kaempferol 4’-O-rut 0.7±0.05 0.8±0.2 Mu3 - kaempferol 3’-O-rut 2.5±0.1 2.8±0.5 Mu5 - amentoflavone 0.9±0.008 1.1±0.2 Table 1d Compounds of Ocimum graticimum Raw Blanched Abrasion Og1 - vicenin-2 1.5±0.002 1.0±0.06 0.9±0.06 Og2 - caffeic acid 0.5±0.01 0.5±0.03 0.3±0.007 Og3 - rutin 0.2±0.003 0.1±0.001 0.02±0.001 Og4 - luteolin-7-O-glucoside 0.3±0.01 0.2±0.02 nd Og5 - kaempferol 3’-O-rutinoside 0.04±0.006 0.03±0.0 nd Og6 - rosmarinic acid 6.1±0.6 29.8±1.7 1.0±0.2 Og10 - cirsimaritin 0.7±0.05 0.6±0.01 0.5±0.02 Og11 - nevadensin 5.8±0.4 5.0±0.3 4.6±0.2 Table 1a-d: Phenolic compounds in processed and unprocessed leaves. All the data are a mean of three different determinations and are expressed as mg/g (SD) dried leaves. constituents (Mu1-Mu4) was unaltered when compared with those observed for the material collected in rainy season [5]. Evaluation of the phenolic content. In Nigeria several green vegetables, among them Va, Mu, Co and Og, are usually blanched before consumption using either hot water or steam. Often indigenous people apply an abrasion treatment to the fresh leaves to remove part of the juice in order to reduce the bitterness and/or acidity of the plant. Among these selected Nigerian plants, this latter treatment is traditionally applied only to Va and Og and, therefore, in this study manually squeezed leaves 2046 Natural Product Communications Vol. 3 (12) 2008 Salawu Sule et al. extracting the phenolic fraction from the leaves and consequently the blanched vegetable, used as food by the local population, remains a good source of phenolic compounds. 45 40 35 30 25 As highlighted for the unprocessed leaves, the total absence of cichoric acid in the Og extracts after blanching and abrasion was confirmed. Moreover, a peculiar behaviour was revealed for the rosmarinic acid that increased consistently (from 6.1 to 29.7 mg/g) in the blanched sample (Table 1d). 20 15 10 5 n ra si o ab O g an ch ed ra w O g bl O g si on ab ra Va ch ed ra w Va bl an Va ch ed ra w bl an M u d nc he o o bl a C C M u ra w 0 Figure 1: Comparison of the total phenolic content in all the samples expressed as mg/g dried leaves. Table 2: DPPH radical scavenging activity. DPPH results from hydroalchoolic extracts Og 0.05 mg/mL 82.9 ±1.0 0.1 mg/mL 88.0 ± 0.4 0.15 mg/mL 88.9 ± 1.0 79.0 ±0.4 Va 55.9 ± 3.2 65.7 ±1.9 Mu 53.1 ±1.4 78.4 ±0.2 81.1±1.8 Co 84.2 ±0.1 79.2± 2.2 88.9 ±1.0 were also analyzed. The quantitative distribution of the different phenolic compounds in the processed and unprocessed leaf extracts is summarized in Tables 1a-d and in the histogram of Figure 1. Comparison of the chromatographic profiles of Co obtained from the blanched and raw leaves showed an inversion of the relative contents of the Co1 and Co5 compounds. Moreover, a consistent decrease of the total phenolic amount (from 42.3 to 5.6 mg/g) in the processed leaves was observed (Table 1a and Figure 1). The greater amount of soluble fiber of this plant [15,16] is removed during the blanching process producing a gel that can entrap part of the more soluble phenols released from the leaves. A dramatic decrease in the total phenolic content was observed for Va after blanching and abrasion, suggesting that these compounds are mainly localized in the juice and consequently they are almost completely removed from the vegetable after these treatments. With regard to Mu, as confirmed by the quantitative data (Table 1c), very few changes were observed between the raw and blanched leaves. Effectively the young Mu leaves, differently from the other plants, are traditionally blanched only for a few minutes. This short time of boiling is not efficacious in Antioxidant activity by DPPH test. One of the aims of this study was to make a preliminary evaluation of the antioxidant potency in terms of free radical scavenging of the phenolic fractions obtained from the leafy vegetables of these plants. Most of the antioxidant activities of vegetables and fruits have been established to be related to phenolic compounds [17]. Free radical scavenging is one of the known mechanisms by which antioxidants inhibit lipid peroxidation [18,19]. The DPPH radical scavenging activity has also been used extensively for screening antioxidants from fruits and vegetables [20]. This activity was measured for the extracts from unprocessed leaves at three different concentrations. A summary of the results expressed as % DPPH radical scavenging activity is reported in Table 2. Overall, all the tested vegetable materials showed a relatively high inhibition at the highest tested concentration and this activity could be attributed to the presence of flavonoids and cinnamoyl derivatives in the hydroalcoholic extracts. The results from Va and Mu seem to be dose dependent. Taking into account the lower concentration, the best results in terms of antioxidant potency were obtained for Og and Co, but no differences among the extracts were highlighted with the higher concentrations. Experimental Materials: The vegetables were harvested from the teaching and research farm of the Federal University of Technology, Akure, Nigeria in March 2008 and voucher specimens were deposited at the Department of Biochemistry, Federal University of Technology, Akure, Nigeria and the Department of Pharmaceutical Science, University of Florence, Italy. About 500 g of fresh green leafy vegetables, Vernonia amygdalina (Va), Corchorous olitorius (Co), Ocimum graticimum (Og) and Manihot utilissima (Mu), were rinsed in water, and the edible portions separated. The edible portions were chopped Phenolic profiles of four tropical green leafy vegetables Natural Product Communications Vol. 3 (12) 2008 2047 into small pieces (300 g) and divided into two (Mu and Co) or three portions (Va and Og). The first portion of the chopped vegetables served as the unprocessed sample, the second portion was blanched in 300 mL boiling water for 15 min (Va, Og and Co) and 5 min for Mu, while the third portion, with the aid of small quantity of water, was manually squeezed by hand (abrasion) to remove the juice. The blanched and squeezed portions were subsequently drained of water. 330 nm (range 0.038-0.3 mg/mL and r2 of 0.9996) was used to evaluate all the cinnamoyl compounds; luteolin 7-O-glucoside at 330 nm (range 0.11-0.88 mg/mL and r2 of 0.9999) was selected to evaluate all the luteolin and apigenin derivatives, together with nevadensin; rutin at 350 nm (range 0.13-1.02 mg/mL and r2 of 0.9999) was used to quantify all the derivatives of quercetin and kaempherol. All the quantitative data were obtained in triplicate. All samples were dried in an air oven at 30°C prior to analysis and treated as described in the next experimental section. The standards used to confirm the chemical structure of some compounds (Table 1a-d) were purchased from Extrasynthese (Geney-France); rutin was from Sigma-Aldrich (St. Louis, MO-USA). Extraction methods for HPLC/DAD analysis of the processed samples: (1 g each) was extracted with stirring for 2 h in 40 mL (20 mL x 2) of ethanol/water 7:3 (v/v) with water acidified with formic acid (pH 2.5). The samples were centrifuged (4,400 rpm for 10 min) and the supernatant was centrifuged again (12,000 rpm for 8 min) to obtain a clear solution directly analyzed by HPLC/DAD. HPLC/DAD/MS analysis: Analyses were performed using a HP 1100 liquid chromatograph equipped with HP DAD and 1100 MS detectors; the interface was a HP 1100 MSD API-electro spray. All the instruments were from Agilent Technology (Palo Alto, CA, USA). The MS analyses were carried out in negative mode with a fragmentor range between 80-150 V. Method 1. A C12 column, 150 × 4 mm (4μm) Synergi Max (Phenomenex-Torrance CA) maintained at 30°C and equipped with a 10 × 4 mm pre-column of the same phase was used with a flow rate of 0.4 mL min-1. The eluents were H2O acidified to pH 3.2 with formic acid (A) and acetonitrile (B). The following linear solvent gradient was applied: from 95 % A to 85% A in 5 min, to 75% A in 8 min and a plateau of 10 min, to 55% A in 12 min and a plateau of 5 min, to 10% A in 3 min, and a final plateau of 2 min to wash the column. The total time of analysis was 45 min. Quantitative determination: Chlorogenic acid, rutin and luteolin 7-O-glucoside were used for the quantitative evaluation. Three five-point calibration curves were prepared as follows: chlorogenic acid at DPPH assay: A dried sample (1 g each) was extracted, by stirring for 2 h, with 40 mL (20 mL x 2) of either ethanol or ethanol/water 7:3 (v/v), with water acidified with formic acid (pH 2.5). The samples were filtered, concentrated to dryness and redissolved in 96% ethanol. The clear solutions were directly analyzed by HPLC/DAD and 3 concentrations (0.05, 0.1 and 0.15 mg/mL) used for the DPPH assay. The radical scavenging activity of ethanol and ethanol/water extracts was carried out as follows: 2 mL of each extract was mixed with 1mL of 0.125 mM DPPH ethanol solution. After shaking the mixture, the absorbance was measured at 517 nm after 5 min of incubation. Radical scavenging activity is expressed as the inhibition percentage. Conclusions: For the first time, seasonal variation in the phenolic content of the four investigated vegetables was highlighted. Within the four plants, almost all showed only quantitative differences, with the exception of Ocimum graticimum. In fact, cichoric acid, previously detected as one of the main constituents of this vegetable collected in November (dry season), was completely absent in the sample harvested in March. The phenolic constituents are chemically unmodified after a strong heating process such as the traditional blanching (about 15 minutes) applied by Nigerian people prior to consuming these vegetables. Nevertheless, these typical preparations showed a consistent decrease in the total phenolic compounds with respect to the raw material, particularly Corchorus olitorius (from 42.3 to 5.56 mg/g dried leaves) and for Vernonia amygdalina (from 40.2 to 4.4 mg/g dried leaves). As expected, when the blanched treatment is reduced to a few minutes, as for Manihot utilissima leaves, the cooked vegetable maintained almost unaltered its original phenolic content (around 10 mg/g dried leaves). The unique exception is the blanched Ocimum gratissimum sample that showed a consistent increment in total phenols, particularly of rosmarinic acid (from 6.1 to 29.8 mg/g dried leaves) in comparison with the unprocessed vegetable. 2048 Natural Product Communications Vol. 3 (12) 2008 Acknowledgments - This research was partially supported by the Italian M.I.U.R. (Ministero Istruzione Università e Ricerca) and we are grateful to Ente Cassa di Risparmio di Firenze for supplying a Salawu Sule et al. part of the instrumentation used for this research. We wish to equally acknowledge the ICTP/IAEA who supported the stay of S.O. Salawu in Italy through a PhD Sandwich Training Educational fellowship. References [1] Grayer RJ, Kite GC, Veitch Nigel C, Eckert MR, Marin PD, Senanayake P, Paton AJ. (2002) Leaf flavonoid glycosides as chemosystematic characters in Ocimum, Biochemical Systematics and Ecology, 30, 327-342. [2] Grayer RJ, Kite GC, Abou-Zaid M, Archer LJ. (2000) The application of atmospheric pressure chemical ionisation liquid chromatography-mass spectrometry in the chemotaxonomic study of flavonoids: Characterisation of flavonoids from Ocimum gratissimum L.var gratissimum. Phytochemical Analysis, 11, 257-267. [3] Adebolu TT, Salau AO. (2005) Antimicrobial activity of leaf extracts of Ocimum gratissimum L.on selected bacteria causing diarrhea in southwester Nigeria. African Journal of Biotechnology, 4, 682-684. [4] Awoyinka AF, Abegunde VO, Adewusi SRA. (1995) Nutrient content of young cassava leaves and assessment of their acceptance as a green leafy vegetable in Nigeria. Plant Foods for Human Nutrition, 47, 21-28. [5] Salawu S. O. Giaccherini C, Innocenti M, Vincieri FF, Akindahunsi AA, Mulinacci N. (2008) HPLC/DAD/MS characterization of flavonoids and cinnamoyl derivatives from some Nigerian green-leafy vegetables. Food Chemistry, accepted. [6] Igile OG, Oleszek W, Jurzysta M, Burda S, Fafunso M, Fasanmide AA. (1994) Flavonoids from Vernonia amygdalina L.and their antioxidant Activities. Journal of Agricultural and Food Chemistry, 42, 2445-2448. [7] Abos AO, Raseroka BH. (2003) In vivo antimalarial activity of Vernonia amygdalina. British Journal of Biomedical Science, 60, 89-91. [8] Masaba SC. (2000) The antimalarial activity of Vernonia amygdalina L. (Compositae).Transactions of the Royal Society of Tropical Medicine and Hygene, 94, 694-695. [9] Oboh G. (2006) Nutritive value and haemolytic properties (in vitro) of the leaves of Vernonia amygdalina L.on human erythrocyte. Nutrition and Health, 182, 151-160. [10] Akinpelu DA. (1999) Antimicrobial activity of Vernonia amygdalina L. leaves. Fitoterapia, 70, 432-434. [11] Erasto P, Grierson AJ, Afolayan AJ. (2006) Bioactive sequiterpene lactones from the leaves of Vernonia amygdalina. Journal of Ethnopharmacology, 106, 117-120. [12] Izevbigie FB. (2003) Discovery of water-soluble anticancer agents (edotides) from a vegetable found in Benin City Nigeria. Experimental Biology and Medicine (Maywood), 228, 293-298. [13] Sakakibara H, Honda Y, Nakagawa S, Ashida H, Kanazawa K. (2003) Simultaneous determination of all polyphenols in vegetables, fruits, and tea. Journal of Agricultural and Food Chemistry, 51, 571-581. [14] Aiyeloja AA, Bello OA. (2006) Ethnobotanical potentials of common herbs in Nigeria: a case study of Enugu State. Educational Research and Review, 1, 16-22. [15] Yamazaki E, Murakami K, Kurita O. (2005) Easy preparation of dietary fiber with the high water-holding capacity from food sources. Plant Foods Human Nutrition, 60, 17-23. [16] Yamazaki E, Kurita O, Matsumura Y. (2008) Hydrocolloid from leaves of Corchorus olitorius L. and its synergistic effect on κ-carrageenan gel strength. Food Hydrocolloids, 22, 819-825. [17] Rice-Evans C, Miller NJ, Bolwell PG, Bramley PM, Pridham JB. (1995) The relative antioxidant activities of plant derived polyphenolic flavonoids. Free Radical Research, 22, 375-383. [18] Rice-Evans C, Miller NJ, Paganga G. (1997) Antioxidant properties of phenolic compounds. Trends in Plant Science, 2, 152-159. [19] Bloknina O, Virolainen E, Fagerstedt KV. (2003) Antioxidants, oxidative damage and oxygen deprivation stress: a review. Annals of Botany, 91, 179-194. [20] Sanchez-Moreno C (2002) Methods used to evaluate the free radical scavenging activities in foods and biological systems. Food Science and Technology International, 8, 121-137. NPC Natural Product Communications (Bio)Sensor Approach in the Evaluation of Polyphenols in Vegetal Matrices 2008 Vol. 3 No. 12 2049 - 2060 M. Camilla Bergonzia*, Maria Minunnib and Anna Rita Biliaa a Dipartimento di Scienze Farmaceutiche, via U. Schiff 6, 50019 Sesto Fiorentino, Firenze, Italy b Dipartimento di Chimica, via della Lastruccia 3, 50019 Sesto Fiorentino, Firenze, Italy mc.bergonzi@unifi.it Received: August 11th, 2008; October 20th, 2008 Polyphenols are compounds widely distributed in the plant kingdom and have attracted much attention, because of their health benefits and important properties such as radical scavenging, metal chelating agents, inhibitors of lipoprotein oxidation, anti-inflammatory and anti-allergic activities. Due to their important role in the diet and in therapy, it is important to estimate their content in the different matrices of interest. Besides classical analytical methods, new emerging technologies have also appeared in the last decade aiming for simple and eventually cheap detection of polyphenols. This review focused on the recent applications of biosensing-based technologies for polyphenol estimation in vegetal matrices, using different transduction principles. These analytical tools are generally fast, giving responses in the order of a few seconds/minutes, and also very sensitive and generally selective (mainly depending on the enzyme used). Direct measurements in most of the investigated matrices were possible, both in aqueous and organic phases. Keywords: Sensor, polyphenols, tyrosinase, laccase, peroxidase, plant tissue, vegetal matrices. Polyphenols are widely distributed in the plant kingdom, including foods of vegetable origin, contributing to their taste and sensorial properties (such as olive oil and wine) and constituting an important role in human diet. These compounds are mainly represented by simple phenolic acids (hydroxybenzoic acids and hydroxycinnamic acids), flavonoids and tannins (hydrolyzable tannins and condensed tannins) [1,2]. Recently, polyphenols have attracted much attention, because of their health benefits, being considered responsible for the majority of the antioxidant capacity in plant-derived products, with a few exceptions, such as carotenoids. It is reported that within the Mediterranean diet, the average daily intake of polyphenols is about 1 g, which is almost 10-fold the intake of vitamin C, 100-fold the intake of vitamin E, and 500-fold the intake of carotenoids [3]. Foods and beverages rich in polyphenols may have a large potential with respect to prevention of diseases, and fruits and vegetables are generally associated with the prevention of stroke [4] and cancers [5,6], including breast cancer [7,8]. Polyphenols act as free radical scavengers [9,10], metal chelating agents [11], inhibitors of lipoprotein oxidation [12] anti-inflammatory agents [13] and have anti-allergic properties [14]. Due to their important role in food and in therapy, it is important that there is a simple and fast estimation of their content in the different matrices of interest. Several methods for polyphenols detection in plant sources are described in the literature, the most known and simplest approach being the FolinCiocalteu spectrophotometric method [15], but this presents limitations since it can estimate other different reducing non-phenolic compounds too. As an alternative to the Folin-Ciocalteu assay, a fluorimetric evaluation of the total phenol content in vegetal matrices and extracts was also proposed [16]. Other classic techniques used in the evaluation of polyphenols are UV-visible [17] and FT-NIR spectroscopies [18]. One of the most selective analytical methods is based on high-performance liquid chromatography (HPLC) combined with different detection methods: UV-Vis detection [19], chemiluminescence detection [20], fluorescence detection, DAD-ESI-MS detection, and 2050 Natural Product Communications Vol. 3 (12) 2008 direct electrochemical detection [21]. However, these instrumental methods, although performing complete and sensitive analyses, require long sample processing (i.e. extraction, concentration, resuspension), massive use of organic solvents, costly instrumentation and skilled personnel. In view of the need for simple and easy analytical methods for rapid estimation of polyphenolic content, one interesting approach is the electroanalytical techniques, due to the electrochemical behaviour shown by phenolic compounds. New emerging technologies have also appeared in the last decade aiming to provide simple, fast and eventually cheap detection of polyphenols. These are mainly based on electrochemical techniques, but some optical-based sensing has also been proposed. In this review, some sensing approaches based on different transduction principles are reported, focusing on polyphenol characterization and quantification. A chemical sensor is a device responding to a chemical stimulus, giving a recordable signal. When the sensor is coupled to a biological molecule, a biosensor is generated. Following the IUPAC definition, “A biosensor is a whole and integrated device providing analytical information (qualitative or semi/quantitative) by using a biomolecular recognition element (biochemical receptor) in close spatial contact with a transducer. The transducer converts the chemical event into a recordable signal. Both sensors and biosensors can be considered innovative analytical devices able to detect different analytes in a qualitative and quantitative manner directly in complex matrices, without or with very little sample pre-treatment. They have been presented eventually as the “analyst dream” for the simplification they make in the analysis [22]. The paradigmatic example is represented by the glucose sensor, which represented a real revolution for diabetes patients; with this very small device (dimension of a pen, also called “glucose pen” in some versions) the glucose content in blood is estimated in a few seconds. The detection principle is amperometry, meaning that a current is the nature of the response, correlated to glucose concentration, and the selectivity is due to the enzyme glucose oxidase, which selectively uses glucose as substrate. The catalysis is at the base of the enzymatic event and for these reasons enzymatic based sensors are called also catalytic sensors. We can thus say that we could Bergonzi et al. Enzymatic (catalytic) The biological element (enzyme) converts the substrate into a product S P Affinity The biological element (receptor) binds specificallythe analyte leading to a complex A+B AB The transducerreveals S or P The transducer reveals the complex Figure1: Catalytic and affinity-based biosensors. discriminate between mainly two different classes of biosensors: catalytic and affinity–based ones (Figure1). The recognition element, immobilized on the sensing surface (i.e. an electrode, an optic fiber, a planar waveguide) is respectively an enzyme (or a system of enzymes) or a receptor able to form an affinity complex with the target analyte. To the affinity sensor category belongs: immunosensors, where the receptors are an antigen or an antibody, or a DNA sensor with suitable probes immobilized on the sensor surface. In Figure 2 are shown some examples of receptors employed both in catalytic and affinity sensors. Relative to polyphenol analysis, catalytic biosensors are the most used. Different enzymes have been used over the years as a catalytic element coupled to electrochemical analysis, such as tyrosinase (also called polyphenol oxidase), laccase and peroxidase, using different electrode materials, flow systems and sample pre-treatment techniques since phenolic compounds can act as electron donors for these enzymes [23-26]. Phenol oxidases and peroxidases have different enzymatic mechanisms of action in the electrochemical biosensors. Enzyme molecules at the surface of the electrode are oxidized by oxygen (for phenol oxidases) or hydrogen peroxide (for peroxidase), followed by their re-reduction by phenolic compounds. The tyrosinase biosensors are restricted to the monitoring of phenolic compounds with at least one free ortho-position. On the other hand, the laccase biosensor can detect free para- and meta-positions, but its catalytic cycle is complicated and in its major part is different from tyrosinase and still now not well understood. Peroxidases exhibit low specificity for electron donors as the phenolic compounds and can be used for phenol detection with certain selectivity and sensitivity. Tyrosinase: The catalytic sensors reported in the literature use mostly the enzyme tyrosinase. The Sensors and polyphenols Natural Product Communications Vol. 3 (12) 2008 2051 Enzyme/Substrate b Antibody/Antigen c Whole cells, tissue d Lipid Layer / Gas e A T G C T A DNA fragments a b c d Signal a e A T G T C A B A Figure 2: Examples of receptors. enzyme catalyzes the oxidation of the phenolic substrate to a quinonic form that is reduced at the electrode polarized at a fixed potential. In the presence of oxygen, this enzyme is capable of catalyzing ortho-hydroxylations of monophenols and oxidation of the consequent ortho-dihydroxyphenols to ortho-quinones. Measurements can be carried out by recording the signal variation related to the dissolved oxygen consumption or to the formation of the relative quinone. Among electroanalytical techniques, in particular, voltammetry has been successfully employed to detect phenols in water media [27-29]. Behaviour of the tyrosinase enzyme electrode has been investigated under different experimental conditions [30-33]. Several authors [34-39] have tested the performance of the tyrosinase electrode in different organic solvents and the effect of different additives has also been investigated [40,41]. Organic phase enzyme electrodes constitute a new class of biosensor applicable to the analysis of substrates or matrices insoluble or scarcely soluble in aqueous media. Many biosensors have a limited lifetime due to enzyme inactivation by the bio-catalytically generated quinone products. For this reason, many studies were made concerning the development of good immobilization methods and materials to improve the biosensor stability. Recently, several amperometric biosensors based on the immobilization of tyrosinase on different electrode materials have been described in the literature. Glassy carbon electrodes modified with polymers [42], sol–gel materials [43], self-assembled monolayers (SAMs) on gold [44], Clark's electrodes [45,46], reticulated vitreous carbon [47] screenprinted [48] carbon paste [49,50], Nafion® membrane [51], hydrogel [52], conducting polymers [53] and natural material such as chitosan [54], and other composite electrodes [55,56] have been used to prepare tyrosinase electrochemical biosensors. Instead of conventional electrodes presenting the limitation of being poisoned after a certain use, recently screen-printed electrodes have been proposed to evaluate the polyphenol content [57]. Screen-printing technology is used for the production of disposable sensors which are very useful because during the oxidation process a polymeric film is formed on the electrode surface leading to electrode surface “inactivation” (“electrode fouling”), one of the main drawbacks of common graphite-based electrodes. Screen-printed electrodes have also been used for screening natural products using bare graphite [58]. The electrochemical device consists of three independent electrodes placed one next to another to form a rectangle 3 cm high and 1.5 cm wide comprising a screen-printed graphite working electrode, a silver reference, and a counter-electrode. Different compounds, including flavones, flavonols, catechins, tannins, and phenylpropanoids were tested with this system. Calibration was performed in a range between 20 and 80 µM of catechin. This method can be useful for a rapid and sensitive screening for polyphenols in plant matrices from grape, olive and green tea. Thus, many applications of biosensors, with different techniques of enzyme immobilization have been concerned with the determination of polyphenols in wine. The use of gold nanoparticles is playing an increasingly important role for the preparation of biosensors [59] and recently, Sanz and coworkers reported the preparation of a tyrosinase biosensor based on the use of a glassy carbon electrode 2052 Natural Product Communications Vol. 3 (12) 2008 modified with electrodeposited gold nanoparticles [57]. The enzyme, immobilized by cross-linking with glutaraldehyde, retains a high bioactivity on this electrode material, giving rise to fast, stable and sensitive responses to various phenolic compounds. The biosensor was applied to the amperometric estimation of the total content of phenolic compounds in red and white wines, which is of interest because of the correlation between wines’ antioxidant capacity and their polyphenol content. The method used an extremely simple procedure involving the direct addition of a sample aliquot to the electrochemical cell. The response was given in a few seconds; the polyphenol concentration found in the wine samples ranged from 16 mg L-1 to 50 mg L-1, expressed as caffeic acid. There was good correlation when the biosensor data were plotted versus the results achieved with the Folin–Ciocalteau method. A different way to entrap the enzyme using electropolymerizing polymers was also reported [6062]. In particular, Böyükbayram and coworkers [62] prepared graft copolymers by electropolymerization of pyrrole with thiophene capped polytetrahydrofuran and used these conducting copolymers to immobilize tyrosinase. The enzyme electrodes were used to determine the amount of phenolic compounds in two brands of Turkish red wine and found very useful owing to their high kinetic parameters and wide pH working range. Thiophene functionalized menthyl monomer with pyrrole (MM/ppy/Tyrosinase) represented another copolymer employed to immobilize tyrosinase [63]. Immobilization of enzyme was performed via entrapment in conducting copolymers during electrochemical polymerization of pyrrole. Maximum reaction rates, Michaelis–Menten constants and temperature, pH and operational stabilities of enzyme electrodes were investigated by the authors. The application of this sensor was again to evaluate the total amount of phenolic compounds in red wines. The immobilized enzyme was optimized at pH 9. MM/ppy/tyrosinase electrode showed stability up to 80°C, while free tyrosinase had an optimal temperature of 40°C and lost its activity completely at 50°C. The system was able to detect the total phenolic compounds present in two different red wines in a range of 3.3-6.0 g/L, expressed as gallic acid equivalents. Different applications reported for tyrosinase deal with the development of artificial senses, such as electronic tongues, where the systems array of sensors with different selectivities are generally integrated to give a response “mimicking” the natural Bergonzi et al. counterpart. A lot is known about artificial ‘nose’, but in the last years some work has appeared dealing with taste. In this regard, Gutés and coworkers [64] elaborated a simultaneous determination of different phenols (phenol, catechol, m-cresol), combining biosensor measurements with chemometric tools and artificial Neural Networks (ANN) analysis. Concentrations of the three phenols ranged from 0 to 130 μM for phenol, 0 to 100 μM for catechol and 0 to 200 μM for m-cresol. As the recognition-detection part, and working electrode, a tyrosinase-based biosensor was developed. The biosensor employs the concept of a graphite-epoxy biocomposite with bulk incorporation of enzyme. Tyrosinase sensors are also employed for quantitative analyses of polyphenols in beer samples. Three amperometric biosensors are described based on immobilization of tyrosinase on a new Sonogel– Carbon electrode for detection of phenols and polyphenols [65]. The electrode was prepared using high energy ultrasound directly applied to the precursors. The first biosensor was obtained by simple adsorption of the enzyme on the Sonogel– Carbon electrode. The second and third ones, presenting sandwich configurations, were initially prepared by adsorption of the enzyme and then modified by mean of a polymeric membrane, such as polyethylene glycol for the second one, and the ionexchanger Nafion in the case of the third biosensor. The optimal enzyme loading and polymer concentration, in the second layer, were found to be 285 U and 0.5%, respectively. All biosensors showed optimal activity under the following conditions: pH 7, −200 mV, and 0.02 mol L−1 phosphate buffer. Sensing performances and kinetic characterizations of the developed biosensors were investigated using some phenolic compounds (catechol, phenol, 4chlorophenol, gallic acid, catechin). In the same paper, the tyrosinase-based Nafion modified Sonogel-Carbon electrode was used to quantify the polyphenol and phenol content of four beers (two lagers and two black) and four environmental water samples [65]. The same research group [66] produced a biosensor based on the bi-immobilization of laccase and tyrosinase. The biosensor employed as the electrochemical transducer the Sonogel-Carbon electrode. The immobilization step was accomplished by doping the electrode surface with a mixture of the enzymes, glutaricdialdehyde and Nafion-ion exchanger, as protective additive. The response of Sensors and polyphenols Natural Product Communications Vol. 3 (12) 2008 2053 this biosensor, carrying Trametes versicolor laccase (Lac) and Mushroom tyrosinase (Ty) based on Sonogel-Carbon detection, was optimized directly in beer samples and its analytical performance with respect to five individual polyphenols was evaluated. The electrode responds to nanomolar concentrations of flavan-3-ols, hydroxycinnamic acids and hydroxybenzoic acids. The limit of detection, sensitivity and linear range for caffeic acid, taken as an example, were 26 nM, 167.53 nA M-1, and 0.01-2 μM, respectively. The Lac-Ty/sonogel-carbon electrode was stable in this matrix, maintaining 80% of its stable response for at least three weeks (RSD 3.6%). The biosensor was applied to estimate the total polyphenol index in ten beer samples and a correlation of 0.99 was obtained when the results were compared with those obtained using the FolinCiocalteau reagent. polyphenols in black tea samples. Enzyme membrane fouling was observed with a number of analyses with a single immobilized enzyme membrane. The tyrosinase-based biosensor gave maximum response to tea polyphenols at 30°C. The optimum pH was 7.0. This biosensor system can be applied in evaluating tea polyphenols quality. Amphiphilic, tyrosinase-modified screen-printed carbon bioelectrodes were decrypted by Cummings [67] for the analysis of lager beers and were compared to the p-dimethylaminocinnamaldehyde (DAC) colorimetric method. Initially, the performances of the biosensors under flowing conditions were appraised using catechol as a model substrate. The electrodes displayed rapid response times and a high degree of sensitivity and reproducibility upon injection of catechol onto a single mainfold. In addition, simple flavanols, separated from barley, were utilized to assess the sensitivity of the biosensors afforded by the presence of the enzyme. The bioelectrode sensitivity decreased upon an increase in molecule size. Finally, using flow injection analysis, authentic beer samples were analyzed and compared to the DAC colorimetric method. A good correlation between the two methods of analysis was observed but, due to the lack of enzyme substrate specificity, the biosensor response did not decrease to the same extent as the colorimetric method; this can be attributed to the presence of interferents, for example, ferulic acid and p-coumaric acid. An amperometric tyrosinase biosensor has also been used for detection of polyphenols in tea [68]. The system could detect tea polyphenols in the concentration range 10–80 mmol L−1. Immobilization of the enzyme, by the crosslinking method, gave a good stable response to tea polyphenols. The biosensor response reached the steady state within 5 min. The voltage response was found to have a direct linear relationship with the concentration of Campo Dall’Orto and coworkers have reported the polyphenol content, expressed as chlorogenic acid equivalents, in a variety of commercially available samples of yerba mate (Ilex paraguaiensis). The compounds were detected using a tyrosinase biosensor and comparing the results with a colorimetric method [69]. The 48% of analyzed samples presented a 92 ± 8 mg of extracted chlorogenic acid equivalents per gram of sample. The extracted chlorogenic acid, expressed as mg/g-1, was evaluated by three methods in a unique yerba mate sample: biosensor (89.2 mg/g-1), Folin (90.2 mg/g-1), and HPLC analysis (21.0 mg/g-1). Biosensing system validation was performed. Repetitiveness of genuine replicates was consistent with the nature of the samples. Discrimination between yerba mate and other plants can be made using principal component analysis (PCA) and the corresponding physical and chemical descriptors. Flavor and taste alterations can be studied by means of analytical methods that involve low-cost instrumentation. Peroxidase: Horseradish peroxidase (HRP) has been eventually employed as another useful enzyme for polyphenol detection. In this approach, the reduction current of oxidized polyphenols, formed during the enzymatic oxidation of polyphenolic compounds in the presence of H2O2, is proportional to their concentration. In other words, the polyphenol content can be detected as the reduction current of the oxidized polyphenol generated by the enzyme reaction cycle of HRP with H2O2. The sensitivity of the detection of various polyphenols by the present method depends on both the electron-donating properties of polyphenols and the electron-accepting properties of oxidized polyphenols. With this approach, Imabayashi [70] reports about the development of a sensor using horseradish peroxidise covalently immobilized on a selfassembled monolayer of mercaptopropionic acid on gold-electrode by the formation of the bond between amino groups on the HRP surface and carboxylic groups on the self-assembled monolayer. The electrode allows polyphenol detection down to 2 μM with a linear relationship up to 25 μM in standard 2054 Natural Product Communications Vol. 3 (12) 2008 solutions. The reduction current of oxidized polyphenols, formed during the enzymatic oxidation of polyphenolic compounds in the presence of H2O2, is proportional to their concentration. The sensitivity of the detection of various polyphenols by the present method depends on both the electron-donating properties of polyphenols and the electron-accepting properties of oxidized polyphenols. When applied to real matrices, such as wine and tea, the total amounts of polyphenols, in the order of μM, were estimated, correlating well with the results determined by the Folin-Ciocalteu method. In another paper [71], the decreased amount of H2O2 caused by the action of peroxidase was sensitively detected with a semipermeable-membrane-covered, HRP-entrapped, and ferrocene-embedded carbon paste electrode. This electrode allows the detection of (+)-catechin down to 0.3 µM and the response is linear up to 15 µM. The same linearity was obtained with other polyphenols found in wine and green tea, such as (-)-epicatechin, caffeic acid, tannic acid and gallic acid. The content of total phenolic compounds in wine [1-8 mM using (+)-catechin as the standard; 2-14 mM using gallic acid as the standard] and tea samples [1-3.5 mM using (+)-catechin as the standard; 0.6-6 mM using gallic acid as the standard] determined by the present method agrees well with results obtained by the Folin-Ciocalteu method. The same enzyme has also been employed immobilized onto silica–titanium and it was applied to measure the polyphenol content of a plant extract without sample pretreatment, because no significant influence of the matrix was observed [72]. Silicabased materials have received greatest interest because they provide a suitable way for designing electrochemical biosensors. Among the silicacontaining matrices, the silica gel modified with metal oxides has been used, not only to improve its amperometric detection, as increase in the internal electrical conductivity of the silica matrix, but also by providing a material with high chemical stability. A biosensor based on horseradish peroxidase and DNA immobilized onto silica–titanium is applied for measuring the polyphenol compounds in plant samples. In the study, various analytical parameters influencing the biosensor performance, such as working potential, type and concentration of the buffer, pH, response time and response in the presence of other compounds, have been investigated as a function of chlorogenic acid (CGA). In the optimized conditions, the biosensor presented a linear Bergonzi et al. response range for CGA from 1 to 50 μmol L−1, applying a potential of −50 mV versus Ag/AgCl, with a sensitivity of 181 μmol−1 L nA cm−2 and detection limit of 0.7 μmol L−1. The biosensor was used to determine the polyphenol content of extracts of coffee and mate. The experimental results showed good agreement with those from the Folin– Ciocalteau method. The polyphenol content in the aqueous extracts of coffee and mate tea ranged from 1.0 to 3.6 (mmol L-1)g-1 of sample. Phenolic compounds are also important factors to be considered in order to evaluate the quality of an extra-virgin olive oil since they are partly responsible for its auto-oxidation stability and organoleptic characteristics. The phenolic content is correlated with many quality parameters, such as the oxidation level or free fatty acidity. Free fatty acids provide an index of the degree of lipase activity and can produce undesirable aromas in the oil; a high value for free fatty acid content indicates a high degree of lipase activity and hence a reduced antioxidant content. Moreover, the oxidation level is dependent upon the composition of the oil and, therefore, upon the degree of unsaturation and the presence of antioxidants, such as phenols. Free fatty acids are responsible for undesirable aromas in the oil. Thus, estimating polyphenol content could provide some indication of oil quality. On the basis of this, some work dealing with olive oil as matrices for polyphenol content evaluation has been reported [73]. Monitoring the polyphenol content (oleuropein derivatives) in an extra-virgin olive oil with varying storage time and storage conditions was performed using two rapid procedures based on disposable screen-printed sensors (SPE) for differential pulse voltammetric analysis, and on an amperometric tyrosinase based biosensor operating in an organic solvent (n-hexane) and using an amperometric oxygen probe as transducer. Differential Pulse Voltammetry (DPV) parameters were chosen in order to study the oxidation of oleuropein, which was used as reference compound. A calibration curve of oleuropein was determined in glycine buffer [10 mM, pH = 2, NaCl 10 mM (D.L. = 0.25 ppm oleuropein, RSD= 7%]. In the case of the tyrosinase based biosensor, the calibration curves were realized using flow injection analysis with phenol as the substrate (detection limit = 4.0 ppm phenol, RSD = 2%). Both of these methods are easy to operate, require no extraction (biosensor) or rapid extraction procedure (Solid Phase Extraction, SPE) and the analysis time is short (min). The results were Sensors and polyphenols comparable with those using reagent and by HPLC analysis. Natural Product Communications Vol. 3 (12) 2008 2055 Folin-Ciocalteau Campanella and coworkers [37] monitored the rancidification process of extra-virgin olive oil using a biosensor operating in organic solvent. The progressive rancidification of the oil was monitored by simultaneously using two different indicators: the peroxide number and an innovative one consisting of the progressive decrease in the content of polyphenols, the main natural antioxidants contained in the oil, as determined rapidly by means of a new organic phase enzyme electrode based on tyrosinase. The aim of the paper was to evaluate the ‘genuineness' of the oil itself and then, above all, to check the correlation between the stability of an olive oil to an artificially induced process of rancidification. The main result of the research was to demonstrate the possibility of using the organic phase enzyme electrode based on tyrosinase to monitor the rancidification process occurring in any sample of olive oil. Indeed, a clear inverse correlation was found throughout the entire oxidation process between the classic indicator, namely the peroxide number, and the polyphenol content of the sample. The simplicity, together with the accuracy and precision of the polyphenol content measurements performed on the olive oil samples revealed the advantages offered by this biosensor. The total polyphenol content in various olive oils ranged from 15.3 to 114.2 mg kg−1 of oil, expressed as phenol. Very recently, the thermal oxidative degradation process of polyphenols was studied, both in a synthetic mixture of five of the more readily available polyphenols contained in the extra-virgin olive oil (EVOO) (tyrosol, vanillin, caffeic acid, ferulic acid and oleuropein) dissolved in glyceryl trioleate, and commercial extra-virgin olive oil [74]. To this end, a series of oxidative degradation experiments was carried out on extra-virgin olive oil samples under isothermal conditions at 98, 120, 140, 160, and 180°C using a thermostatic silicon oil bath. The change in polyphenol concentration with time was monitored at selected temperatures using a tyrosinase biosensor operating in an organic phase (nhexane). The EVOO rancidification process rate displayed good inverse correlation between the variation in the peroxide value, the more traditional index, and that of a more innovative index determined by the concentration of the “total polyphenols” (expressed in mol L−1 of phenol). In this paper, the authors analyzed the kinetic degradation process and the kinetic parameters of the process were determined through an isothermal study carried out at different temperatures (between 98 and 180°C). Laccase: Some studies reported the use of laccase as a possible enzyme for development of biosensors for phenols and polyphenols. Laccase, a coppercontaining oxidase, is widely distributed in fungi, higher plants and in some bacteria. The use of a laccase-modified electrode for detection of flavonoids was reported by Gorton’s group [75]. In a recent study, the laccase from Cerrena unicolor, as a highly active enzyme, coupled to amperometric transduction was reported for the detection of flavonoids. The enzyme was adsorptively immobilized on the surface of a graphite electrode. In particular, catechin hydrate, epicatechin, epicatechin gallate, prodelphinidin, and caffeic acid were used as target compounds. Electrodes modified with laccase yield responses for both simple compounds and compounds with three or more phenolic and nonphenolic rings, but with different sensitivities. Considering wine as a matrix of interest, another example of a laccase biosensor is given. A biosensor developed with Laccase Coriolus versicolor immobilized on derivatized polyethersulfone membranes and applied to a Pt–Ag, AgCl US electrode base was applied to evaluate several polyphenols usually found in red wine (caffeic acid, gallic acid, catechin, rutin, trans-resveratrol, quercetin and malvidin) [76,77]. It was observed that an amperometric response was obtained for catechin at +100 mV (versus Ag/AgCl) and caffeic acid at −50 mV in acetate buffer solutions (pH 4.5) having 12% ethanol. At pH 3.5 and +100 mV the biosensor was sensitive to both substrates and their response was additive. One limit of this biosensor is the necessity for a previous solid phase extraction of the matrix for polyphenol enrichment; large interferences can occur. Amperometric determination using a biosensor based on immobilized laccase was applied for the analysis of tannins of tea at different stages of its production [78]. The enzymes were from Coriolus versicolor, Coriolus hirsutus and Cerrena maxima, and immobilized on threadlike DEAE-cellulose. The time needed for analysis in the flow injection mode was below 100 s. A column with immobilized enzyme could be used for up to 500 determinations of phenolic compounds (tannin content 100-199 mg/g of dry substance) without decrease of the enzyme activity. 2056 Natural Product Communications Vol. 3 (12) 2008 The use of a laccase biosensor, both under batch and flow injection conditions, for a rapid and reliable amperometric estimation of the total content of polyphenolic compounds in wines is also reported [79]. The enzyme was immobilized by cross-linking with glutaraldehyde onto a glassy carbon electrode. Caffeic acid and gallic acid were selected as standard compounds to carry out such an estimation. Experimental variables, such as the enzyme loading, the applied potential, and the pH value, were optimized, and different aspects regarding the operational stability of the laccase biosensor were evaluated. Using batch amperometry at -200 mV, the detection limits obtained were 2.6 × 10-3 and 7.2 × 10-4 mg L-1 gallic acid and caffeic acid, respectively, which compares advantageously with previous biosensor designs. An extremely simple sample treatment consisting only of an appropriate dilution of the wine sample with the supporting electrolyte solution (0.1 mol L-1 citrate buffer of pH 5.0) was needed for the amperometric analysis of red, rosé, and white wines. Good correlations were found when the polyphenol indices obtained with the biosensor (in both the batch and FI modes) for different wine samples were plotted versus the results achieved with the classic Folin-Ciocalteu method. Application of the calibration transfer chemometric model (multiplicative fitting) allowed for the confidence intervals (for a significance level of 0.05) for the slope and intercept values of the amperometric index versus the Folin-Ciocalteu index plots (r = 0.997) including the unit and zero values, respectively. This indicates that the laccase biosensor can be successfully used for the estimation of the polyphenol index of wines and is comparable with the FolinCiocalteu reference method. Plant tissue: An alterative approach to the use of purified enzymes is the employment of whole tissue which contains different enzymes sets. Whole tissue materials from plants or animals provide many advantages for the construction of biosensors. In some cases, plant tissue containing polyphenol oxidase (e.g., banana, potato, apple, and burdock) has been used, coupled to electrodes, for the detection of catechol-related components, such as flavonols and catechins in beers and green tea [78,80-83]. The linear detection range of the plant-tissue electrodes, depending on the enzyme preparation used, was, on average, between 2 and 12 µM catechins. Tissue sensors have been applied to the determination of flavonols in beers [81]. In particular, different Bergonzi et al. plant tissues, banana, potato and apple, containing tyrosinase have been evaluated for their ability to detect catechol related components in beers. Calibration graphs were produced for each plant tissue with both catechol and (−)-epicatechin as standards. The response of the banana based sensor was rather erratic and showed a large zero error, probably due to flavanols in the banana. Potato, wet apple and dried apple were all satisfactory. The response with dried apple was 60% that of wet apple, but showed greater stability. The response of (−)epicatechin was 1.4 to 1.5 times that of catechol with either wet or dried apple. Banana and potato based sensors were used for the determination of total flavanols in a range of commercial beers and lagers. Good analytical data were obtained with potato, comparable to those obtained using colorimetric or liquid chromatographic analyses. The best biosensors were from potato and apple. While there was slight loss of response due to the drying process, dried apple has greater longevity and excellent response characteristics. In preliminary experiments reported elsewhere, the same authors [82] have demonstrated that the flavanol components in beer can be determined with a ‘bananatrode’ biosensor based on the carbon paste electrode using catechol as the standard. A burdock (Arctium lappa L., a biennial plant) tissuebased biosensor was applied for measuring total catechins in green tea infusions [83]. This catechin biosensor was found to respond to five catechins (catechin/epicatechin/epigallocatechin/epicatechin gallate/epigallocatechin gallate), gallic acid, catechol and ascorbic acid and to total catechins in green tea infusions. The precision of the measurements was good (< 3% RSD) and the biosensor showed no interference from major amino acids or carbohydrates in the infusions. One limitation of this approach, however, is that the biosensor is inadequate for accurate quantitation of total catechins because of the severe variability in the relative biosensor response to the different catechins. Jewell and Ebeler developed a simple tyrosinasebased biosensor composed of 5% banana tissue, 10% mineral oil, and 85% carbon-containing ruthenium, mixed together in a small beaker to form a stiff paste, for the measurement in a winery or food setting for rapid and simple phenolics detection [84]. This was achieved by the design and construction of an operational amplifier-based tyrosinase biosensor. Excellent correlation was shown between the biosensor and the Folin-Ciocalteu assay for simple Sensors and polyphenols Natural Product Communications Vol. 3 (12) 2008 2057 phenolics (gallic acid, catechin, epicatechin, caffeic acid, quercetin, seed tannins) and for wines. The simple compounds and the seed tannins were chosen in order to examine their biosensor response, in correlation with different structure, OH groups and complexity of matrix. The varying signals observed were due to the chemical structure and variable number of OH groups associated with polyphenols, which caused different reaction with tyrosinase; this is also observed with all redox methods. No interferences were observed in a model wine solution made according to standard wine phenolic concentrations due to low pH, tartaric acid or ethanol present in the model wine. Finally, one white wine and three red wines, selected to examine the effects of aging and the different phenolic profiles, were analyzed with the biosensor. Also in this case, the response between Folin-Ciocalteau and biosensor methods was similar, and oak aging and varietal difference did not appreciably influence the biosensor response [84]. was also demonstrated on the examples of red and white wines [85]. Behind electrochemical detection, optical sensing also represents an interesting feature for phenols detection. One particular application of this sensor was reported by Edelmann and Lendl, who developed an electronic tongue to evaluate the tannin content of red wine [85]. The interaction of tannins with proline-rich proteins (gelatin) was studied using an automated flow injection system with Fourier transform infrared spectroscopic detection to gain insight into chemical aspects related to astringency. The PRP gelatin was selected to mimic the parotid salivary proteins. In the perception of astringency, an interaction between proline-rich salivary proteins and tannins present in the sample takes place. To study this interaction, agarose beads carrying gelatin (a proline-rich protein) were placed in the IR flow cell in such a way that the beads were probed by the IR beam. Using an automated flow system, samples were injected in a carrier stream and flushed over the proteins in a highly reproducible manner. Simultaneously, any retardation due to tannin-protein interactions taking place inside the flow cell were monitored by infrared spectroscopy. Tannins of different sources (grapes, wooden barrels, formulations used in wine making) were investigated, and their flow-through behaviour was characterized. Significant differences in their affinity toward gelatin could be observed. Furthermore, because of small but characteristic differences in the IR spectrum, it is possible to distinguish condensed from hydrolysable tannins. The selectivity of the flow-through sensor Concluding remarks: A review of the literature concerning the application of biosensor technology to vegetal matrices has been reported and discussed. Special attention has been focused on the use of these devices for the identification and quantification of polyphenols and, in some cases, for the quality control of these matrices. Usually the determination of total polyphenol content is performed by spectrophotometric or chromatographic methods, but in recent years, with the aim to develop methods capable of being employed also in situ, several biosensors have been developed to determine phenols in either aqueous or organic solvents. Organic phase enzyme electrodes constitute a new class of biosensor applicable to the analysis of substrates or matrices insoluble or scarcely soluble in aqueous media. Many different sensors have been developed in the last 15 years for polyphenol detection and electrochemical transduction is the approach most applied. Different enzymes have been used over the years as the catalytic element coupled to electrochemical analysis, such as tyrosinase, laccase, and peroxidase, since phenolic compounds can act as electron donors for these enzymes. In some cases, plant tissue containing polyphenol oxidase (e.g., banana, potato, apple and burdock) has also been used for the detection of polyphenols in vegetal matrices. The mostly investigated vegetal matrices are those with a great commercial interest such as olive oil, wine, beer and tea. By the use of screen-printed electrodes, different phenols, including flavones, flavonols, catechins, tannins, and phenylpropanoids were tested with these analytical tools. Some limitations occur with these devices in real matrices, such as the working conditions (pH, temperature) and the risk of enzyme inactivation. Many biosensors have a limited lifetime due to enzyme inactivation by the biocatalytically generated quinone products. For this reason, many studies were concerned with the development of good immobilization methods and materials to improve the biosensor stability. Recently, the immobilization of enzymes in electropolymerized conducting polymers, sol–gel materials, gold nanoparticles, Clark's electrodes, screen-printed, carbon paste, hydrogels, has received a great deal of interest. 2058 Natural Product Communications Vol. 3 (12) 2008 Another limitation of this approach, however, is that sometime the biosensor is inadequate for accurate quantification of total polyphenols because of the severe variability in the relative biosensor response to the different phenol derivatives. Nevertheless, biosensors may provide a promising competitive technology for a simple, fast and sensitive detection of polyphenolic compounds without any pre-treatment. In most of the cases successfully employed for the analysis of real Bergonzi et al. samples, comparable results were obtained with conventional analysis by HPLC. Thus, the impact of biosensors in food and plant matrices in the future will be enormous because they are a label-free screening system, which imparts flexibility to the process of assay design and facilitates successful integration with other technologies thanks to the increasing number of commercially available instruments having novel sensor surfaces, immobilization techniques and attachment chemistries. References [1] [2] [3] [4] [5] [6] [7] [8] [9] [10] [11] [12] [13] [14] [15] [16] [17] [18] [19] [20] [21] [22] [23] [24] [25] Herrmann K. (1989) Occurrence and content of hydroxycinnamic and hydroxybenzoic acid compounds in foods. Critical Reviews in Food Science and Nutrition, 28, 315-347. Escarpa A, Gonzalez MC. (2001) An overview of analytical chemistry of phenolic compounds in foods. Critical Reviews in Analytical Chemistry, 31, 57-139. Scalbert A, Williamson G. (2000) Dietary intake and bioavailability of polyphenols. Journal of Nutrition, 130, 2073S-2085S. Ness AR, Powles JW. (1997) Fruit and vegetables, and cardiovascular disease: a review. International Journal of Epidemiology, 26, 1-13. Steinmetz KA, Potter JD. (1996) Vegetables, fruit, and cancer prevention: a review. Journal of American Dietetic Association, 96, 1027-1039. Huang MT, Ferraro T. (1992) Phenolic-compounds in food and cancer prevention, ACS Symposium Series 507, p. 8. Jansen MC, McKenna D, Bueno-De-Mesquita HB, Feskens EJ, Streppel MT, Kok FJ, Kromhout D. (2004) Reports: Quantity and variety of fruit and vegetable consumption and cancer risk. Nutrition and Cancer, 48, 142-148. Lamartiniere CA. (2000) Protection against breast cancer with genistein: a component of soy. American Journal of Clinical Nutrition, 71, 1705S-1709S. Halliwell B, Aeschbach R, Löliger J, Aruoma OI. (1995) The characterization of antioxidants. Food Chemistry and Toxicology, 33, 601-617. Rice Evans CA, Miller J, Paganga G. (1997) Antioxidant properties of phenolic compounds. Trends in Plant Science, 2, 152-159. Haslam E. (1996) Natural polyphenols as drugs: possible modes of action, Journal of Natural Products, 59, 205-215. Salvi A, Brihlmann C, Migliavacca E, Carrupt PA, Hostettmann K, Testa B. (2002) Protein protection by antioxidants: developments of a convenient assay and structure–activity relationships of natural polyphenols. Helvetica Chimica Acta, 85, 867–881. Read MA. (1995) Flavonoids-naturally occurring anti-inflammatory agents. American Journal of Pathology. 147, 235-237. Cody V, Middleton Jr E, Harborne JB, Beretz A, Alan R. (1988) Plant Flavonoids in Biology and Medicine. II. Biochemical, Cellular and Medicinal Properties. In Progress in Clinical and Biological Research, vol. 280, Liss, New York. Cunniff PA. (1995) Official Methods of Analysis, AOAC International, Arlington. Papoti VT, Tsimidou MZ. (2009) Looking through the qualities of a fluorimetric assay for the total phenol content estimation in virgin olive oil, olive fruit or leaf polar extract. Food Chemistry, 112, 246-252. Dao L, Friedman M. (1992) Chlorogenic acid content of fresh and processed potatoes determined by ultraviolet spectrophotometry. Journal of Agricultural and Food Chemistry, 40, 2152-2156 Zhao QCJ, Cai MLJ, Liu J. (2008) Determination of total polyphenols content in green tea using FT-NIR spectroscopy and different PLS algorithms. Journal of Pharmaceutical and Biomedical Analysis, 46, 568-573. Simón BF, Pérez-Ilzarbe J, Hernández T, Goméz-Cordovés C, Estrella I. (1992) Importance of phenolic compounds for the characterization of fruit juices. Journal of Agriculural Food Chemistry, 40, 1531-1535. Ogawa A, Arai H, Tanizawa T, Miyahara T, Toyo’oka T. (1999) On-line screening method for antioxidants by liquidchromatography with chemiluminescence detection. Analytica Chimica Acta, 383, 221-230. Escarpa C, Gonzalez MC. (2001) An overview of analytical chemistry of phenolic compounds in foods. Critical Reviews in Analytical Chemistry, 31, 57-139. Turner APF, Karube I, Wilson GS. (1987) Biosensor- Fundamentals and Applications, 1st Ed., Oxford University Press. Haghighi B, Gorton L, Ruzgas T, Jonsson LJ. (2003) Characterization of graphite electrodes modified with laccase from Trametes versicolor and their use for bioelectrochemical monitoring of phenolic compounds in flow-injection analysis. Analytica Chimica Acta, 487, 3-14. Freire RS, Duran N, Kubota LT. (2001) Effects of fungal laccase immobilization procedures for the development of a biosensor for phenol compounds. Talanta, 54, 681-686. Yaropolov I, Kharybin AN, Emneus J, Marko-Varga G, Gorton L. (1995) Flow-injection analysis of phenols at a graphite electrode modified with co-immobilized laccase and tyrosinase. Analytica Chimica Acta, 308, 137-144, Sensors and polyphenols [26] [27] [28] [29] [30] [31] [32] [33] [34] [35] [36] [37] [38] [39] [40] [41] [42] [43] [44] [45] [46] [47] [48] [49] [50] [51] [52] [53] [54] [55] [56] Natural Product Communications Vol. 3 (12) 2008 2059 Lindgren L, Stoica T, Ruzgas A, Ciucu F, Gorton L. (1999) Development of a cellobiose dehydrogenase modified electrode for amperometric detection of diphenols. Analyst, 124, 527-532. Campanella L, Beone T, Sammartino MP, Tomassetti M. (1993) Determination of phenol in waste water using an enzyme sensor. Analyst, 118, 979-986. Lindgren A, Ruzgas T, Emneus J, Csoregi E, Gorton L, Marko Varga G. (1996) Flow injection analysis of phenolic compound with carbon paste electrodes modified with tyrosinase purchase from different companies. Analytical Letters, 29, 1055-1068. Mazzei F, Botrè F, Lanzi M, Lorenti G, Porcelli F, Botrè C. (1992) Plant metabolism as an analytical tool: some applications of plant tissue electrodes for the selective determination of catecholamines. Sensors and Actuators B, 7, 427-430. Campanella L, Favero G, Sammartino MP, Tomassetti M. (1994) The effect of organic solvent properties on the response of a tyrosinase enzyme sensor. Talanta, 41, 1015-1023. Onnerfjord PK, Emnéus J, Marko-Varga G, Gorton L, Ortega F, Domínguez E. (1995) Tyrosinase graphite-epoxy based composite electrodes for detection of phenols. Biosensors and Bioelectronics, 10, 607-619. Marko-Varga G, Emneus J, Gorton L. (1995) Development of enzyme-based amperometric sensors for the determination of phenolic compounds. Trends in Analytical Chemistry, 14, 319-328. Nistor C., Emnéus J., Gorton L., Ciucu A. (1999) Improved stability and altered selectivity of tyrosinase based graphite electrodes for detection of phenolic compounds. Analytica Chimica Acta, 387, 309-326. Iwuoha EI, Adeyoju O, Dempsey E, Smyth MR, Liu J, Wang J. (1995) Investigation of the effects of polar organic solvents on the activity of tyrosinase entrapped in a poly(ester-sulphonic acid) polymer. Biosensors and Bioelectronics, 10, 661-667. Capannesi C, Palchetti I, Mascini M, Parenti A. (2000) Electrochemical sensor and biosensor for polyphenols detection in olive oils. Food Chemistry, 71, 553-562 Wang J, Naser N, Kwon HS, Cho MY. (1992) Tissue bioelectrode for organic-phase enzymatic assays. Analytica Chimica Acta, 264, 7-12. Campanella L, Favero G, Pastorino M, Tomassetti M. (1999) Monitoring the rancidification process in olive oils using operating in organic solvents, Biosensensors and Bioelectronics, 14, 179-186. Campanella L, Fortuney A, Sammartino MP, Tomassetti M. (1994) Tyrosinase biosensor response as a function of physical properties of organic solvents. Talanta, 41, 1397-1404. Campanella L, Favero G, Sammartino MP, Tomassetti M. (1998) The effect of organic solvent properties on the response of a tyrosinase enzyme sensor. Talanta, 46, 595-606. Lutz M, Burestedt E, Emnéus J, Lidén H, Gobhadi S, Gorton L, Marko-Varga G. (1995) Effect of different additives on a tyrosinase based carbon paste electrode. Analytica Chimica Acta, 305, 8-17. Campanella L, Favero G, Persi L, Sammartino MP, Tomassetti M, Visco G. (2001) Organic phase enzyme electrodes: applications and theoretical studies. Analytica Chimica Acta, 426, 235-247. Védrine C, Fabiano S, Tran-Minh C. (2003) Amperometric tyrosinase based biosensor using an electrogenerated polythiophene film as an entrapment support. Talanta, 59, 535-544. Yu J, Liu S, Ju H. (2003) Mediator-free phenol sensor based on titania sol–gel encapsulation matrix for immobilization of tyrosinase by a vapor deposition method. Biosensors and Bioelectronics, 19, 509-514. Campuzano S, Serra B, Pedrero M, de Villena FJM, Pingarrón JM. (2003) Amperometric flow-injection determination of phenolic compounds at self-assembled monolayer-based tyrosinase biosensors. Analytica Chimica Acta, 494, 187-197. Streffer K, Vijgenboom E, Tepper AWJW, Makower A, Scheller FW, Canters GW, Wollenberger U. (2001) Analytica Chimica Acta, 427, 201-210. Hasebe Y, Yokobori K, Fukasawa K, Kogure T, Uchiyama S. (1997) Highly sensitive electrochemical determination of Escherichia coli density using tyrosinase-based chemically amplified biosensor Analytica Chimica Acta, 357, 51-54. Peña N, Reviejo AJ, Pingarrón JM. (2001) Detection of phenolic compounds in flow systems based on tyrosinase-modified reticulated vitreous carbon electrodes. Talanta, 55, 179-187. Chang SC, Rawson K, McNeil CJ. (2002) Disposable tyrosinase-peroxidase bi-enzyme sensor for amperometric detection of phenols Biosensors and Bioelectronics, 17, 1015-1023. Liu S, Yu J, Ju H. (2003) Renewable phenol biosensor based on a tyrosinase-colloidal gold modified carbon paste electrode Journal of Electroanalytical Chemistry, 540, 61-67. Rogers KR, Becker JY, Cembrano J. (2000) Improved selective electrocatalytic oxidation of phenols by tyrosinase-based carbon paste electrode biosensor. Electrochimica Acta, 45, 4373-4379. Furbee J, Thomas C, Kelly R, Malachowski M. (1993) Mediated electrochemical reduction of cytochrome C and tyrosinase at perfluorosulfonated ionomer coated electrodes. Analytical Chemistry, 65, 1654-1657. Daigle F, Leech D. (1997) Reagentless tyrosinase enzyme electrodes: Effects of enzyme loading, electrolyte pH, ionic strength, and temperature. Analytical Chemistry, 69, 4108-4112. Adeloju SB, Shaw SJ. (1993) Polypyrrole-based potentiometric biosensor for urea part 1. Incorporation of urease. Analytical Chimica Acta, 281, 611-615. Edwards W, Leukes WD, Rose PD, Burton SG. (1999) Immobilization of polyphenol oxidase on chitosan-coated polysulphone capillary membranes for improved phenolic effluent bioremediation Enzyymatic Microbiology Technology, 25, 769-773. Serra B, Jiménez S, Mena L, Reviejo J, Pingarrón JM. (2002) Composite electrochemical biosensors: a comparison of three different electrode matrices for the construction of amperometric tyrosinase biosensors. Biosensors and Bioelectronics, 17, 217-226. Önnerfjord P, Emnéus J, Marko-Varga G, Gorton L, Ortega F, Domínguez E. (1995) Tyrosinase graphite-epoxy based composite electrodes for detection of phenols Biosensors and Bioelectronics, 10, 607-619. 2060 Natural Product Communications Vol. 3 (12) 2008 [57] [58] [59] [60] [61] [62] [63] [64] [65] [66] [67] [68] [69] [70] [71] [72] [73] [74] [75] [76] [77] [78] [79] [80] [81] [82] [83] [84] [85] Bergonzi et al. Carralero Sanz V, Luz Mena M, González-Cortés A, Yáñez-Sedeño P, Pingarrón JM. (2005) Development of a tyrosinase biosensor based on gold nanoparticles-modified glassy carbon electrodes: Application to the measurement of a bioelectrochemical polyphenols index in wines. Analytica Chimica Acta, 528, 1-8. Romani A, Minunni M, Mulinacci N, Pinelli P, Vincieri FF, Del Carlo M, Mascini M (2000) Comparison among differential pulse voltammetry, amperometric biosensor, and HPLC/DAD analysis for polyphenol determination. Journal of Agricultural and Food Chemistry, 48, 1197-1203. Liu S, Leech D, Ju H. (2003) Application of colloidal gold in protein immobilization, electron transfer, and biosensing. Analytical Letters, 36, 1-19. Cosnier S. (1999) Biomolecule immobilization on electrode surfaces by entrapment for attachment to electrochemically polymerized films. A review. Biosensors & Bioelectronics, 14, 443-456. Cosnier S. (2003) Biosensors based on electropolymerized films: new trends. Analytical and Bioanalytical Chemistry, 377, 507-520. Elif Böyükbayram A, Kıralp S, Toppare L, Yağcı Y. (2006) Preparation of biosensors by immobilization of polyphenol oxidase in conducting copolymers and their use in determination of phenolic compounds in red wine. Bioelectrochemistry, 69, 164-171. Kıralp S, Toppare L, Yagcı Y. (2003) Immobilization of polyphenol oxidase in conducting copolymers and determination of phenolic compounds in wines with enzyme electrodes. International Journal of Biological Macromolecules, 33, 37-41. Gutés A, Céspedes F, Alegret S, del Valle M. (2005) Determination of phenolic compounds by a polyphenol oxidase amperometric biosensor and artificial neural network analysis. Biosensors and Bioelectronics, 20, 1668-1673. El Kaoutit M, Naranjo-Rodriguez I, Temsamani KR, Hidalgo-Hidalgo de Cisneros JL. (2007) The Sonogel–Carbon materials as basis for development of enzyme biosensors for phenols and polyphenols monitoring: A detailed comparative study of three immobilization matrixes. Biosensors and Bioelectronics, 22, 2958-2966. El Kautit M, Naranjo-Rodriguez I, Temsamani KR, de la Vega MD, Hidalgo-Hidalgo de Cisneros JL. (2007) Dual Laccasetyrosinase based Sonogel-Carbon biosensor for monitorung polyphenols in beers. Journal of Agricultural and Food Chemistry, 55, 8011-8018. Cummings EA, Eggins BR, McAdams ET, Linquette-Mailley S, Mailley P, Madigan D, Clements M, Coleman C. (2001) Development of a tyrosinase-based, screen-printed amperometric electrode for the detection of flavanoid polyphenols in lager beers. Journal of American Society of Brewing Chemists, 59, 84-89. Abhijith KS, Sujith Kumar PV, Kumar M.A, Thakur MS. (2007) Immobilised tyrosinase-based biosensor for the detection of tea polyphenols. Analytical and Bioanalytical Chemistry, 389, 2227-2234. Campo Dall’Orto V, Vago JM, Carballo RR, Rezzano IN. (2005) Comparison of tyrosinase biosensor and colorimetric method for polyphenol analysis in different kinds of teas. Analytical Letters, 38, 19-33. Imabayashi SI, Kong YT, Watanabe M. (2001) Amperometric biosensor of polyphenols based on horseradish peroxidise immobilized on gold electrodes. Electroanalysis, 13, 408-412. Kong YT, Imabayashi SI, Kano K, Ikeda T, Kakiuchi T. (2001) Peroxidase-based amperometric sensor for the determination of total phenols using two-stage peroxidase reactions. American Journal of Enology and Viticulture, 52, 381-385. Mello LD, Del Pilar Taboada Sotomayor M, Kubota LT. (2003) HRP-based amperometric biosensor for the polyphenols determination in vegetables extract. Sensors and Actuators B: Chemical, 96, 636-645. Capannesi C, Palchetti I, Mascini M, Parenti A. (2000) Electrochemical sensor and biosensor for polyphenols detection in olive oils. Food Chemistry, 71, 553-562. Campanella L, Nuccilli A, Tomassetti M, Vecchio S. (2008) Biosensor analysis for the kinetic study of polyphenols deterioration during the forced thermal oxidation of extra-virgin olive oil. Talanta, 74, 1287-1298 Jarosz-Wilko azka A, Ruzgas T, Gorton L. (2004) Use of laccase-modified electrode for amperometric detection of plant flavonoids. Enzyme and Microbial Technology, 35, 238-241. Gomes SASS, Nogueira JMF, Rebelo MJF. (2004) An amperometric biosensor for polyphenolic compounds in red wine. Biosensors and Bioelectronics, 20, 1211-1216. Gomes SASS, Nogueira JMF, Rebelo MJF. (2003) A new laccase biosensor for polyphenols determination. Sensors, 3, 166-175. Ghindilis AL, Gavrilova VP, Yaropolov AI. (1992) Laccase-based biosensor for determination of polyphenols: determination of catechols in tea. Biosensors & Bioelectronics, 7, 127-131. Gamella M, Campuzano S, Reviejo AJ, Pingarrón JM. (2006) Electrochemical estimation of the polyphenol index in wines using a laccase biosensor. Journal of Agricultural and Food Chemistry, 54, 7960–7967. Botrè F. (1991) Plant tissue electrode for the determination of catechol. Analytica Chimica Acta, 255, 59-64. Eggins BR, Hickey C, Toft SA, Min Zhou D. (1997) Determination of flavanols in beers with tissue biosensors Analytica Chimica Acta, 347, 281-288 Eggins BR. (1996) Biosensors: An Introduction, Wiley-Teubner,Chichester-Stuttgart p. 179. Horie H, Mukai T, Goto T, Kawanaka M, Shimohara T. (1994) Measurement of tea catechins using biosensors. Nippon Shokuhin Kogyo Gakkaishi, 41, 433-435. Jewell WT, Ebeler SE. (2001) Tyrosinase biosensor for the measurement of wine polyphenolics. American Journal of Enolology and Viticulture, 52, 219-222. Edelmann A, Lendl B. (2002) Toward the optical tongue: flow-through sensing of tannin-protein interactions based on FTIR spectroscopy. Journal of American Chemical Society, 124, 14741-14747. NPC Natural Product Communications In vitro Radical Scavenging and Anti-Yeast Activity of Extracts from Leaves of Aloe Species Growing in Congo 2008 Vol. 3 No. 12 2061 - 2064 Annalisa Romania, Pamela Vignolinia, Laura Isolanib, Sara Tombellic, Daniela Heimlerb, Benedetta Turchettid and Pietro Buzzinid ,* a Dipartimento di Scienze Farmaceutiche, Università degli Studi di Firenze, I-50019 Sesto Fiorentino, Italy b Dipartimento di Scienza del Suolo e Nutrizione della Pianta, Università degli Studi di Firenze, I-50144 Firenze, Italy c Dipartimento di Chimica, Università degli Studi di Firenze, I-50019 Sesto Fiorentino, Italy d Dipartimento di Biologia Applicata, Sezione di Microbiologia, Università di Perugia, I-06100 Perugia, Italy pbuzzini@unipg.it Received: September 8th, 2008; Accepted: October 29th, 2008 Extracts obtained from leaves of Aloe barbadensis and A. congolensis, growing in Congo, were analyzed for their in vitro antiradical and anti-yeast activity. Different leaf tissues (tegument and gel) were analyzed separately. Their phenolic fractions showed the presence of chromones and anthrones (aloesin, aloin B, aloin A, and isoaloeresin), flavonoids (apigenin and kaempferol derivatives), and hydroxycinnamic acids. A differential quantitative composition was observed between leaf tegument and gel: in the first, higher concentrations of the four classes of compounds were observed. The extracts from the tegument exhibited higher in vitro antiradical and antimycotic activity than gel extracts. In a few cases, extracts from teguments were active against amphotericin B-insensitive yeasts. Due to the lack of radical scavenging and yeast inhibition observed when aloin was used, it was possible to postulate that the in vitro activities of the teguments could be related to their high concentration of flavonoids and hydroxycinnamic acids. Keywords: Aloe spp., antiradical activity, anti-yeast activity, polyphenols, DPPH• radical, yeasts. Biological activities (e.g. antioxidant, antiradical, anticarcinogenic, antimutagenic, antiproliferative and antimicrobial) expressed by extracts obtained from plant tissues are well known [1]. The increasing interest in discovering novel plant extracts exhibiting biological activities is justified by their potential role in supporting or even substituting commercial drugs. The genus Aloe L. (Liliaceae) includes over 300 species of perennial herbs, shrubs and trees. Among them, A. barbadensis Miller (trivially labeled as A. vera) is a perennial herb with rosettes of long pointed leaves coming from a shortly branched, creeping rhizome. The leaves are brittle and exude a clear yellowish viscous sap when broken. Although it is widespread throughout the African continent, this species is considered to originate from southern and eastern Africa and from the Mediterranean regions. Leaves of A. barbadensis are characterized by a high water content and more than 60% of the dry weight is composed of polysaccharides [2]. Phenolics of the anthrone and chromone type have been described as the main secondary metabolites [3,4]. A. congolensis, which grows in a broad area of Congo, received its first botanical description in 1899 [5]. Neither the phenolic pattern nor the biological activity of this species has been reported. Since antiquity, gel obtained from the succulent leaves of A. barbadensis has been used in folk medicine for obtaining preparations (traditionally known as “medicinal aloe”) for treatment of internal and external diseases, both in humans and animals [6]; this has been extensively proposed as a bactericidal and fungicidal drug [7], as well as for dermatological applications, especially for radiationcaused skin lesions [8,9]. However, no evidence has 2062 Natural Product Communications Vol. 3 (12) 2008 Romani et al. so far been published on the antiradical and anti-yeast activity of extracts from teguments of Aloe spp. Thus the aim of the present study was the characterization of the phenolic composition of extracts of the teguments and gel of A. barbadensis and A. congolensis, and the in vitro determination of their antiradical and anti-yeast activity (against species implicated in human mycoses). Table 1: General characteristics of Aloe leaves. Data are mean values of ten determinations (standard deviation within brackets). Chromones, anthrones, flavonoids and hydroxycinnamic acids were detected in chromatograms of the phenolic fractions of extracts of A. barbadensis and A. congolensis teguments and gels. Chromones and anthrones have previously been detected in the phenolic fractions of Aloe leaves [3,10], whereas no previous studies have reported the presence of flavonoids and hydroxycinnamic acids. On the basis of their UV-Vis and mass spectra, apigenin diglucoside and three kaempferol derivatives were detected in A. barbadensis leaves. The most representative compounds in the tegument extract of this species were aloesin, aloin B, aloin A, and isoaloeresin (the last two compounds not being separated), which were also observed in the gel. On the contrary, flavonoids and hydroxycinnamic acids were detected exclusively in the tegument. Table 2: Quantitative data obtained from HPLC measurements. All data are expressed as mg/g fresh weight. Standard deviations within brackets. Kinetic curves of the in vitro antiradical activity of extracts from A. barbadensis and A. congolensis teguments and gels are reported in Figure 1. In the first case, a higher antiradical activity has been observed. This evidence is in agreement with results obtained by Hu and co-workers [11]. In addition, tegument of A. congolensis exhibited a higher antiradical activity than that of A. barbadensis (80.7% vs 41.3%) (Figure 1). Since in preliminary tests we found that the aloin standard gave negative results in the DPPH test (unpublished data), we might Length cm Weight g Gel % A. barbadensis 27 (3.5) 61.9 (7.9) 55.7 23.6 3.2 A. congolensis 26 (4.2) 34.1 (6.2) 32.0 33.2 5.0 Samples Total anthrone and chromone Total flavonoids A. barbadensis teguments A. barbadensis gel A. congolensis teguments A. congolensis gel 2.32 (0.35) 0.06 (0.01) 2.50 (0.29) 0.03 (0.01) 0.11 (0.003) 0.004 (0.001) 0.53 (0.064) 0.009 (0.002) Total hydroxycinnamic derivatives 0.014 (0.006) traces 0.042 (0.097) traces 0.3 a) 0.25 b) absorbance (nm) By comparing the HPLC profile of A. congolensis with that of A. barbadensis, it was possible to observe that the composition of the tegument was characterized by a lesser amount of phenolics; in particular, an apigenin derivative is one of the main compounds. On the contrary, the gel of A. congolensis exhibited a higher amount of phenolics than that of A. barbadensis. It should, however, be pointed out that the amount of gel obtained is lower in the case of A. congolensis (see Table 1). Quantitative data of total phenolics, obtained from the chromatograms and calculated on the basis of aloin, apigenin-7-O-glucoside, and caffeic acid calibration curves, are reported in Table 2. Yield of lyophilised sample mg/g Tegument Gel Species 0.2 c) 0.15 0.1 d) 0.05 0 0 5 10 15 20 25 30 35 time (min) Figure 1: Kinetic curves of the reaction with the stable DPPH• radical of : a) gel barbadensis (2 g/mL); b) gel congolensis (2 g/mL); c) tegument barbadensis (3 g/mL); d) tegument congolensis (3 g/mL) suppose that the observed antiradical activity could be related to the presence of flavonoids and hydroxycinnamic acids (5.2% and 18.7% of the total polyphenol content for A. barbadensis and A. congolensis, respectively) (Table 2). Extracts of teguments of both species exhibited a broad antimycotic activity against Candida albicans, C. glabrata, C. tropicalis, Clavispora lusitaniae (former Candida lusitaniae), Issatchenkia orientalis (teleomorph of Candida krusei), Filobasidiella neoformans (former Cryptococcus neoformans) and Pichia guilliermondii (former Candida giulliermondii) (Table 3). Interestingly, in a few cases, extracts of teguments were active against Antiradical and anti-yeast activity of Aloe spp. Natural Product Communications Vol. 3 (12) 2008 2063 Table 3: Antimycotic activity of extracts of Aloe barbadensis and A. congolensis. n.a. = no activity; AmB = Amphotericin. DBVPG Species accession number Candida albicans Candida glabrata Candida parapsilosis Candida tropicalis Clavispora lusitaniae Filobasidiella neoformans Issatchenkia orientalis Pichia guilliermondii Diameter of inhibition halos (mm) A. A. barbadensis congolensis tegument tegument 250 mg/mL 250 mg/mL AmB 100 μg/mL 6133 21.8 20.0 22.5 3828 16.0 12.0 18.7 6150 n.a. n.a. n.a. 3982 16.2 12.1 n.a. 6142 14.7 13.0 n.a. 6010 30.5 28.4 15.9 6782 13.2 12.6 n.a. 6140 18.5 17.0 n.a. amphotericin B-insensitive yeast strains (Table 3). As reported above, in vitro antimicrobial activity of extracts of A. barbadensis has been reported previously [7], but, to the best of our knowledge, this is the first report of anti-yeast activity of extracts of teguments of A. congolensis. Likewise with the antiradical activity, as there was a lack of yeast inhibition observed when aloin was used (unpublished data), we might speculate that the in vitro anti-yeast activity of the Aloe species teguments could be related to their high concentration of flavonoids and hydroxycinnamic acids (Table 2). Experimental Chemicals and reagents: Methanol, acetonitrile (HPLC grade), methylene chloride, and formic acid (ACS reagent) were purchased from Aldrich Company Inc. (Milwaukee, Wiscosin), and aloin, aloemodin, caffeic acid and apigenin-7-O-glucoside from Extrasynthèse (Lyon, Nord-Genay, France). Plant materials: Three years-old Aloe plants were sampled at Kimbondo, Mont’ngafula, near Kinshasa (Democratic Republic of Congo) in June 2005. A. barbadensis was cultivated in a nursery in Minkoti, whereas a local ecotype of A. congolensis was sampled in its natural habitat in Minkoti. Living plants were transported to the laboratory for further sampling and analytical procedures. Gel and tegument of succulent leaves were separately extracted with ethanol-water (70:30, pH 3.2 with formic acid) in the dark for 12 h. The extracts were defatted twice with n-hexane, rinsed with water and Table 4: Linear solvent gradient system used in HPLC-DAD and HPLCMS analysis of Aloe samples. Analysis was carried out during a 58 min period at as flow rate of 1 mL/min using a Lichrosorb C18 (250 x 4.6 mm i.d., 5 μm) column operating at 27°C. Time min H2O/H+ % CH3CN % Flow mL/min 0 20 100 85 0 15 1 1 25 85 15 1 35 75 25 1 43 75 25 1 53 0 100 1 58 0 100 1 subsequently lyophilized for 12 h. Length and weight of leaves, weight of gel and the percentage of lyophilized sample are reported in Table 1. The lyophilized extracts were dissolved in ethanol-water (70:30, pH 3.2 with formic acid) and directly analyzed by HPLC/DAD and HPLC/MS. HPLC/DAD and HPLC/MS analysis: The lyophilized extracts were analyzed by reverse-phase and normal-phase high performance liquid chromatography. The analysis was carried out using a HP-1100 liquid chromatograph equipped with a DAD detector and a HP 1100 MSD API-electrospray (Agilent-Technologies, Palo Alto, USA) operating in positive and negative ionization mode. The elution conditions are reported in Table 4. Identification of individual compounds was carried out on the basis of their retention times, spectroscopic and spectrometric data, using aloin, aloe emodin, luteolin-7-Oglucoside and caffeic acid as reference compounds. Calibration curves with r2 ≥ 0.998 were considered. The quantification was performed at the maximum wavelength of UV-Vis absorbance by applying the correction for molecular weight, and the reported values are the means of three determination. Determination of the in vitro antiradical activity: Free radical scavenging activity was evaluated with the DPPH• (1,1-diphenyl-2-picrylhydrazyl radical) assay. The antiradical capacity of lyophilized extracts was estimated according to the procedure reported by Brand-Williams [12] and slightly modified. Two mL of an ethanol solution of lyophilized extracts were added to 2 mL of an ethanol solution of DPPH• (0.0025g/100mL) and the mixture kept at room temperature. The absorption was measured at 517 nm with a Lambda 25 spectrophotometer (Perkin-Elmer) with ethanol as a blank. The percentage of inhibition was calculated according to the following formula: 2064 Natural Product Communications Vol. 3 (12) 2008 %inhibition = [( At =0 − At = 20 ) / At =0 ]× 100 The absorption of the DPPH• solution was checked daily. Determination of in vitro anti-yeast activity: Eight yeast strains (each one representing the type strain of 8 different pathogenic species, belonging to 5 genera) [13-16] were used as target microorganisms. All strains are conserved in the Industrial Yeast Collection DBVPG, University of Perugia, Italy, www.agr.unipg.it/dbvpg. Anti-yeast activity exhibited by lyophilized extracts of A. barbadensis and Romani et al. A. congolensis (as above prepared) were determined by using the agar diffusion well bioassay (ADWB) [16,17]. Amphotericin B (AmB) (Calbiochem Inc., USA) was also tested as a control anti-yeast agent. Acknowledgments – We thank Dr Claudio Aroldi for his technical assistance and Dr Padre Hugo Rios, Pediatric Foundation of Kimbondo, for sample collection. The authors wish to express their sincere gratitude to the Cassa di Risparmio di Firenze that contributed to the acquisition of part of the instrumentation used for this work. References [1] [2] [3] [4] [5] [6] [7] [8] [9] [10] [11] [12] [13] [14] [15] [16] [17] (a) Rusia K, Srivastava SK. (1988) Antimicrobial activity of some Indian medicinal plants. Indian Journal of Pharmaceutical Sciences, Jan-Feb, 57-58; (b) Ayoub SMH. (1989) Antimicrobial screening of Libyan medicinal plants. Planta Medica, 55, 650-651; (c) Mahajan V, Arora DS, Sabherwal U. (1991) Antibacterial activity of some tea samples. Indian Journal of Microbiology, 31, 443-445; (d) Etkin NL. (1996) Medicinal cuisines: diet and ethnopharmacology. International Journal of Pharmacognosy, 34, 313-326; (e) Arora DS, Ohlan D. (1997) In vitro studies of antifungal activity of tea (Camellia sinensis) and coffee (Coffea arabica) against wood rotting fungi. Journal of Basic Microbiology, 24, 127-131; (f) Chapman L, Johns T, Mahunnah RLA. (1997) Saponin-like in vitro characteristics of extracts from selected non-nutrient wild plant food additives used by Maasai in meat and milk based soups. Ecology, Food and Nutrition, 36, 1-22; (g) Cowan MM. (1999) Plant products as antimicrobial agents. Clinical. Microbiology Review, 12, 564-582; (h) Johns T. (1999) Plant constituents and the nutrition and health of indigenous peoples. In Ethnoecology. Situated Knowledge, Located Lives. Nazarena, VD. (Ed). The University of Arizona Press, Tucson, 157-174; (i) Pieroni A. (2000) Medicinal plants and food medicines in the folk traditions of the upper Lucca Province, Italy. Journal of Ethnopharmacology, 70, 235-273; (j) Buzzini P, Pieroni A. (2003) Antimicrobial activity of extracts of Clematis vitalba towards pathogenic yeast and yeast-like microorganisms. Fitoterapia, 74, 397-400; (k) Surh YJ. (2003) Cancer chemoprevention with dietary phytochemicals. Nature Review on Cancer, 3, 768-780; (l) Shahidi F, Naczk M. (2004) Phenolics in Food and Nutraceuticals. CRC Press, Boca Raton, FL, USA; (m) Alasalvar C, Karamacä M, Amarowicz R, Shahidi F. (2006) Antioxidant and antiradical activities in extracts of hazelnut kernel (Corylus avellana L.) and hazelnut green leafy cover. Journal of Agriculture and Food Chemistry, 54, 4826-4832. Femenia A, Sanchez ES, Simal S, Rossellò C. (1999) Compositional features of polysaccharides from Aloe vera (Aloe barbadensis Miller) plant tissues. Carbohydrate Polymers, 39, 109-117. Okamura N, Asai M, Hine N, Yagi A. (1996) High-performance liquid chromatographic determination of phenolic compounds in Aloe species. Journal of Chromatography A, 746, 225-231. Park MK, Park JH, Kim NY, Shin YG, Choi YS, Lee JG, Kim KH, Lee SK. (1998) Analysis of 13 phenolic compounds in Aloe species by high-performance liquid chromatography. Phytochemical Analysis, 9, 186-191. Reynolds GW. (1966) The Aloes of Tropical Africa and Madagascar. Aloes Book Fund Publisher, Swaziland. Reynolds T, Dweck AC. (1999) Aloe vera leaf gel: a review update. Journal of Ethnopharmacology, 68, 3-37. Shelton RM. (1991) Aloe vera. Its chemical and therapeutic properties. International Journal of Dermatology, 30, 679-683. Davis RH, Leitner MG, Russo JM, Byrne ME. (1989) Wound healing. Oral and topic activity of Aloe vera. Journal of American Pediatrics Medical Association, 84, 77-81. Kaufman T, Newman AR, Wexler MR. (1989) Aloe vera and burn wound healing. Plastic Reconstruction Surgery, 83, 1075-1076. Esteban-Carrasco A, Lopez-Serrano M, Zapata JM, Sabater B, Martin M. (2001) Oxidation of phenolic compounds from Aloe barbadensis by peroxidase activity: possible involvement in defense reaction. Plant Physiology and Biochemistry, 39, 521-527. Hu Q, Hu Y, Xu J. (2005) Free radical-scavenging activity of Aloe vera (Aloe barbadensis Miller) extracts by supercritical carbon dioxide extraction. Food Chemistry, 91, 85-90. Brand Williams W, Cuvelier ME. (1995) Use of a free radical method to evaluate the antioxidant activity. Lebensmittel Wissenschaft und Technology, 28, 25-30. Hazen KC. (1995) New and emerging yeast pathogens. Clinical Microbiology Review, 8, 462-478. Rex JH, Rinaldi MG, Pfaller MA. (1995) Resistance of Candida species to fluconazole. Antimicrobial Agents and Chemotherapy, 39, 1-8. Warren NG, Hazen KC. (1998) Candida, Cryptococcus and other yeasts of medical importance. In Manual of Clinical Microbiology. Murray PR, Baron EJ, Pfaller MA, Tenover FC, Yolker RH. (Eds), ASM Press, Washington DC, 1184-1198. Buzzini P, Martini A. (2001) Large-scale screening of selected Candida maltosa, Debaryomyces hansenii and Pichia anomala killer toxin activity against pathogenic yeasts. Medical Mycology, 39, 479-482. Turchetti B, Pinelli P, Buzzini P, Romani A, Heimler D. (2005) In vitro antimycotic activity of some plant extracts towards yeast and yeast-like strains. Phytotherapy Research, 19, 44-49. NPC Natural Product Communications Antioxidant Principles and Volatile Constituents from the North-western Iberian mint “erva-peixeira”, Mentha cervina 2008 Vol. 3 No. 12 2065 - 2068 Matteo Politi, * César L Rodrigues, Maria S Gião, Manuela E Pintado and Paula ML Castro Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Dr. António Bernardino de Almeida, 4200-072, Porto-Portugal mpoliti@esb.ucp.pt Received: July 5th, 2008; Accepted: October 29th, 2008 Total phenol content determined by the Folin–Ciocalteu assay and total antioxidant capacity measured by the ABTS•+ method were applied for the first time to analyze the aqueous extract prepared from the dried aerial parts of Mentha cervina, a Portuguese mint species traditionally used as a culinary herb in river fish-based dishes and currently commercialized to prepare digestive infusions. LC-MS/MS analysis was performed directly on the crude aqueous extract allowing the identification of seven phenolic compounds. The overall plant aroma was analyzed by the SPME/GC-MS method; this approach allowed the characterization of various constituents, as well as the comparison between the fresh and dried plant material. Such a comparison highlighted several metabolic changes that occur during the drying process of the aerial parts of this plant. Keywords: Mentha cervina, traditional recipe, infusion, volatiles, antioxidant, LC-MS, SPME/GC-MS. Mentha cervina (L.) Opiz (Lamiaceae), previously named Preslia cervina (L.) Fresn., is a mint growing wild in stony places and on the banks of rivers in north-western regions of the Iberian peninsula. It is traditionally used as a culinary herb, especially to aromatize river fish-based dishes in Portugal, mainly in the interior regions, such as along the “Sabor” river rather than along the sea coast. The freshly caught fish are wrapped with the fresh leaves and steamed [1a]; this explains its common name “ervapeixeira” that means fisher-herb. Contrary to other mint species, this plant is not so competitive and it is currently difficult to find it growing spontaneously in the natural habitat. The recently renewed interest in this aromatic species has induced local farmers, such as the “Cantinho das Aromáticas” in Porto district and “Ervital” in Viseu district, to cultivate it for selling either as a living plant or as dried material (aerial parts). Generic recommendations for its consumption are about the use of the fresh plant for culinary purpose while the dried material is generally used to prepare infusions with digestive properties. In both cases, the aerial parts of the plants are used. To the best of our knowledge, the only scientific article currently available on this species is by Gonçalves and co-workers [1b], who reported the variation of the essential oil constituents with the seasons and the corresponding in-vitro antifungal activity. The major objective of the present work was the phytochemical investigation, by LC-MS/MS analysis, of the aqueous extract (infusion) prepared from the dried plant, as this extract had not been studied before. Following our intensive investigation of medicinal and food plants currently commercialized in Portugal [2a], specific emphasis was given to total phenol content calculated by the Folin–Ciocalteu assay [2b], and the total antioxidant capacity measured by the ABTS•+ method [2c]. Antioxidants are now increasingly required in the human diet because of their benefits to health and it is, therefore, relevant to evaluate their content within plants sold in the market as food ingredients or medicine. The second part of our investigation describes the analysis of the volatile constituents through the SPME/GC-MS method. Such an approach allowed the rapid metabolic profile of the plant aroma without the need of the essential oil extraction by hydrodistillation, as previously performed by Gonçalves and co-workers [1b]. This approach was used here with the aim to compare the fresh and dried plant material. Such a comparison can highlight the chemical uniqueness of this traditional culinary recipe that indicates the preferential use of the fresh 2066 Natural Product Communications Vol. 3 (12) 2008 “erva-peixeira” to aromatize the Portuguese river fish-based dishes. The antioxidant capacity of M. cervina was measured by the ABTS•+ assay as ascorbic acid equivalents, and the total phenol content was calculated by the Folin–Ciocalteu method as gallic acid equivalents. By comparison of such data with those from 48 other infusions prepared from powdered medicinal plants currently commercialized in Portugal and which had been analyzed using the same assays and extraction procedures [2a], M. cervina presents a medium antioxidant capacity with a value of 0.16 ± 0.013 g L-1 of ascorbic acid equivalents (avocado leaves Persea americana Mill.- was the best antioxidant with 1.43 ± 0.13 g L-1 while the black elder flower Sambucus nigra L.- was the weakest with 0.043 ± 0.003 g L-1). Using the same comparison, M. cervina showed an intermediate total phenol content with a value of 0.15 ± 0.00 g L-1 of gallic acid equivalents (avocado leaves gave a value of 0.55 ± 0.03 g L-1 and black elder flower 0.040 ± 0.007 g L-1). Moreover, M. cervina was less active as an antioxidant compared with Mentha x piperita L. (0.40 ± 0.10 g L1 ), but as good as M. spicata L. (0.14 ± 0.07 g L-1), while the total phenol content was half that of the two other mints (M. x piperita 0.31 ± 0.11 g L-1; M. spicata 0.35 ± 0.03 g L-1). LC-MS/MS analysis was performed directly on the crude aqueous extract (infusion prepared from powdered aerial parts of the plant). The aim of such analysis was to identify as many phenols as possible contained in this extract; normally, the presence of this class of compounds is well related with the antioxidant activity measured by the ABTS•+ method. The identification of such phenols was achieved by comparison with pure standards previously injected using the same chromatographic (LC) and detection (MS/MS) conditions. Our in house library of phenols contains 33 compounds but, from these, no more than 7 were identified in the aqueous extract of M. cervina (Table 1). Among them, chlorogenic and caffeic acids were the most abundant, followed by cumaric acid and rutin. To the best of our knowledge, these constituents are reported for the first time in this mint species. Chlorogenic acid is a well known cholagogue and choleretic derivative [3]; its regular ingestion helps the flow of bile, facilitating, therefore, the digestive process. The relatively high amount of such a Politi et al. Table 1: LC-MS/MS analysis of M. cervina infusion; % values are indicative of the relative amounts of the seven identified compounds calculated by integration of the area under the peaks. Compound name Protocatechuic acid Cumaric acid Caffeic acid Epicatechin Chlorogenic acid Orientin Rutin Retention time (min.) 9.65 20.16 18.30 22.84 16.75 20.50 [M-1]- Fragments 153.0 163.0 179.0 289.0 353.0 447.0 25.52 609.0 109.0 (100) 119.0 (100) 135.0 (100) 245.0 (100) 191.0 (100) 357.0 (70); 327.0 (100); 285.0 (20) 301.0 (40) Relative amount 3.1% 19.7% 29.1% 1.1% 32.1% 3.3% 11.6% constituent in M. cervina aqueous extract is in accord with the claimed digestive properties of this preparation. The characteristic fragrance of aromatic plants is the result of the sum of the single volatile compounds, and their relative percentages are essential to determine their characteristic perfumes. In this work, the use of the SPME/GC-MS method allowed the comparison of the volatile constituents of the fresh and dried M. cervina aerial parts; both materials came from the same plant (see Experimental section). Biochemical changes attributable to the drying process of the plant were detected. Such modification appears to be mostly semi-quantitative rather than qualitative. The area under the peaks in the chromatograms is proportional to the amount of the corresponding compounds. Data on the relative amount (semi-quantitative data) of the single chemical entities were, therefore, obtained by comparison of the area of the corresponding peaks. Such comparison was performed within the same chromatogram as well as between both chromatograms obtained from the fresh and dried material. This was because, as detailed in the Experimental section, the same amount of plant (1 g) was used as starting material in both cases (1 g of fresh aerial parts was directly analyzed, while another 1 g from the same plant was dried before analysis). In accordance with the results obtained by Gonçalves and co-workers [1b] on the essential oil constituents of M. cervina, the major volatiles detected were pulegone, isomenthone, and limonene, among others not identified here. Concerning the major compound pulegone, it was impossible to derive the area under the peak because this compound was so abundant that saturation of the MS detector occurred in both cases, for the fresh and dried plant. The amount of limonene and isomenthone were higher in the dried plant compared with the fresh one; however, the relative amounts of both constituents changed differently. In Phytochemical investigation of Mentha cervina Natural Product Communications Vol. 3 (12) 2008 2067 fact, the amount of limonene was three times higher in the dried plant with respect to the fresh one, while isomenthone was only two times more abundant in the dried material. This indicates that the increase in certain constituents during the drying process of the plant can be preferential with respect to others (in this case limonene augments three times, while isomenthone only two). Other compounds, such as those detected at RT 42.01, 42.92, and 61.44 min were only slightly more abundant in the fresh material rather than in the dried. Other minor differences, in this case truly qualitative, were noted such as, for instance, that a peak at RT 43.70 was only present in the fresh material, and a peak at RT 44.58 min only in the dried plant. From the analysis of the corresponding MS spectra (not shown), both compounds appear to be isomers. Such chemical modifications, which occur during the drying process of the plant, produce a characteristic metabolic fingerprinting of the fresh mint distinguishable from the fingerprinting of the dried plant. This can be considered as a chemical reason for the distinctivness of the Portuguese culinary river fish-based recipe that recommends the use of the fresh mint. used to characterise the Portuguese culinary recipe that recommends the use of the fresh mint. Allowing direct chemical analysis of the volatile constituents without any previous treatment of the plant material, the SPME/GC-MS method is proposed here as the most adequate technique for the study of other aromatic plants used in traditional culinary or medicinal recipes. The overall aroma of the fresh and dried mint vary, not only in terms of intensity (dried plant has stronger aroma compared with the corresponding fresh one), but also in terms of quality. The overall aroma of the fresh mint appears in particular much gentler. Therefore, the fresh plant is possibly more appropriate for the delicate taste of these fish-based dishes. New chemical and biological data on M. cervina are described in this work. Despite the lack of scientific data on the aqueous extract of this mint, the dried plant is currently commercialised in Portugal to prepare an infusion with claimed digestive properties. It was, therefore, our wish to describe its chemical content at least in term of known phenolic compounds with potential antioxidant capacity; the presence of seven phenolic derivatives were here described for the first time in this mint species. Total antioxidant capacity and total phenol content were compared with other commercial medicinal plants currently marketed in Portugal. Concerning the overall aroma analysis of M. cervina, it was possible to detect some chemical differences between the volatile constituents of the fresh and the dried plant material. Such differences were detected by using the SPME/GC-MS approach, here applied for the first time to this mint species. The chemical data acquired on the dried and fresh material of M. cervina can be Experimental Sample preparation: The dried aerial parts of M. cervina were kindly provided by Ervital (Castro Daire, Portugal). The dried plant material was milled prior to the preparation of the infusion that was obtained as follows: 110 mL of boiling water was added to 1 g of powdered plant; after 5 min (i.e. the time period typically used by the consumer) the extract was filtered through a 0.45 µm filter. The fresh and dried aerial parts of M. cervina here compared by SPME/GC-MS analysis were purchased from “Cantinho das Aromáticas” (Vila Nova de Gaia, Portugal). Fresh aerial parts (1 g) were directly analyzed by SPME/GC-MS, while another 1 g from the same plant was dried at room temperature in the shade for 6 days obtaining, at the end, 0.25 g of dried material. Total antioxidant capacity: This activity was measured through the ABTS•+ method, as previously described [2a]. Briefly, the ABTS•+ solution was prepared by addition, at 1:1 (v/v), of 7 mmol L−1 ABTS (2,2-azinobis(3ethylbenzothiazoline-6sulfonic acid) diammonium salt (Sigma-Aldrich)) to 2.45 mmol L−1 potassium persulfate (Merck, Darmstadt, Germany); the reaction took place in the dark for 16 h. In order to obtain an absorbance of 0.700 ± 0.020, at 734 nm, measured with an UV 1203 spectrophotometer (Shimadzu, Tokyo, Japan), the ABTS•+ solution was diluted in ultra-pure water. For analysis of experimental samples, an accurate volume was used in order to obtain an inhibition percentage between 20 and 80%, after 6 min of reaction, with 1 mL of ABTS•+ solution; the average of three replicates was used as a datum point. Total antioxidant capacity was expressed as percentage of inhibition (PI), according to the equation PI = (Abs ABTS•+ − Abs sample)/Abs ABTS•+) × 100, where Abs ABTS•+ denotes the initial absorbance of diluted ABTS•+, and Abs sample denotes the absorbance of the sample after 6 min of reaction. Ascorbic acid (99.0% pure, from Sigma-Aldrich, Steinheim, Germany) was used as a standard. Quantitative results (in g L−1 of ascorbic acid equivalents) were 2068 Natural Product Communications Vol. 3 (12) 2008 obtained through calibration curves produced using standard solutions of ascorbic acid. Using this calibration curve, the final result was thus expressed as an equivalent concentration of ascorbic acid (in g L−1). Total phenol content: The amount of phenolic compounds was determined as described elsewhere [2a,2b]. To 0.5 mL of sample, 0.5 mL of Folin– Ciocalteu reagent (Merck), 10mL of 75 g L−1 sodium carbonate (Sigma-Aldrich) and water were added to a final volume of 25 mL. Absorbance at 750 nm was measured on a Heλios α spectrophotometer (Unicam, Cambridge, UK). Gallic acid was used as a standard to prepare calibration curves in the ranges 4–80 and 20–400 mg L−1. Total phenol content was reported as gallic acid equivalents (C, in g L−1), using the expression C = (Abs sample − 0.0201)/2.1456, where Abs sample denotes absorbance of the sample after 1 h of reaction. The Pearson correlation coefficient of the above fit was 0.9991. LC-MS/MS analysis: The chromatographic system consisted of a Prostar 210 LC pump (Varian, CA, USA) coupled with a Varian 1200 triple quadrupole mass spectrometer (Varian, CA, USA) with electrospray ionization in positive and negative modes. A 5 µm C18 column (4.6mm x100 mm, Merck) was used for the separation at a flow rate of 0.4 mL min-1. The LC-MS/MS method previously developed by Sun et al. [4] was partially modified in this work. LC separation was performed in 30 minutes using a gradient elution (eluent A, water with 0.1% formic acid; eluent B, 100% methanol: time 0 min, A 90%; 12.05 min, A 78%; 22.05 min, A 50%; 27.05 min, A 95%; 30 min A, 95%). ESI-MS/MS detection was obtained using a capillary voltage of 55V. For MS/MS fragmentation, argon atoms were used (pressure 1.20 mtorr; collision energy of 15 V). Data were acquired with a Varian LC-MS 1200L Politi et al. Workstation. An in-house LC-MS/MS library was created by injecting 33 pure phenolic standards using the abovementioned LC-MS/MS conditions. The identification of the phenolic compounds contained in the M. cervina extract was achieved by direct injection of the crude extract and comparison with the in-house library. SPME/GC-MS analysis: The conditions chosen for extraction were as follows: 1 g of fresh and 0.25 g of dried M. cervina were placed in 40 mL flasks, and capped with a gas-tight seal; the samples were gently stirred during volatile collection using a stirring bar, at room temperature, while a divinylbenzene/carboxen/polydimethylsiloxane (DVB/CAR/PDMS) fiber was introduced and left for 10 min to trap the volatiles; afterwards, the SPME fiber was placed in the injector port of a GC-MS Focus GC Thermo Scientific, and left to desorb (trapped) the volatiles for 10 min. The conditions selected for separation were as follows: thermal desorption at the injector port was at 220ºC, in splitless mode, with the split valve being opened 30 s after injection; resolution was through a FFAP column (50 m x 0.22 mm ID BP21, 0.25 μm), with an oven temperature held at 40ºC for 1 min, and then increased at 2ºC min-1 up to 220ºC, which was eventually held for 30 min; helium C-60 (Gasin, Portugal) was used as carrier gas, at a volumetric flow rate of 1 mL min-1. Acknowledgments - The dried plant material was kindly provided by “Ervital”, while the fresh plant was obtained from “Cantinho das Aromáticas”. We express gratitude, in particular to Luís Alves, founder of the “Cantinho das Aromáticas”, for his precious suggestions and continuous encouragement. We thank the European Union (MRTN-CT-2006036053) for financial support through the INSOLEX project. References [1] [2] [3] [4] (a) Proença da Cunha A, Alves Ribeiro J, Rodrigues Roque O. (2007) Plantas Aromáticas em Portugal Caracterização e Utilizações. Fundação Calouste Gulbenkian ed., Lisbon. pp. 1-328; (b) Gonçalves MJ, Vicente AM, Cavaleiro C, Salgueiro L. (2007) Composition and antifungal activity of the essential oil of Mentha cervina from Portugal. Natural Product Research, 21, 867–871. (a) Gião MS, Gonzalez-Sanjose ML, Rivero-Perez MD, Pereira CI, Pintado ME, Malcata FX. (2007) Infusions of Portuguese medicinal plants: Dependence of final antioxidant capacity and phenol content on extraction features. Journal of the Science of Food and Agriculture, 87, 2638–2647; (b) Singleton VL, Rossi JA. (1965) Colorimetry of total phenolics with phosphomolybdic– phosphotungstic acid reagents. American Journal of Enology and Viticulture, 16, 144–158; (c) Re R, Pellegrini N, Proteggente A, Pannala A, Yang M, Rice-Evans C. (1999) Antioxidant activity applying an improved ABTS radical cation decolorization assay. Free Radical Biology and Medicine, 26, 1231–1237. Proença da Cunha A, Pereira Da Silva A, Rodrigues Roque O. (2006) Plantas e Produtos Vegetais em Fitoterapia. Fundação Calouste Gulbenkian ed., Lisbon. pp. 1-702. Sun J, Liang-Bin Y, Li P, Duan C. (2007) Screening non-colored phenolics in red wines using liquid chromatography/ultraviolet and mass spectrometry/mass spectrometry libraries. Molecules, 12, 679-693. NPC Natural Product Communications Chemical Composition of Thymus serrulatus Hochst. ex Benth. Essential Oils from Ethiopia: a Statistical Approach 2008 Vol. 3 No. 12 2069 - 2074 Bruno Tirillinia*, Roberto Maria Pellegrinob, Mario Chessac and Giorgio Pintorec a Institute of Botany, University of Urbino, via Bramante 28, 61029 Urbino, Italy b Department of Chemistry, Laboratory of Organic Chemistry, University of Perugia, via Elce di Sotto 8, 06123 Perugia, Italy c Dip. Farmaco-Chimico-Tossicologico, University of Sassari, Via Muroni 23, I-07100 Sassari, Italy bruno.tirillini@uniurb.it Received: July 22nd, 2008; Accepted: October 17th, 2008 From the essential oil (EO) obtained from the aerial parts of T. serrulatus collected in Ethiopia, fifty-three compounds were identified by GC/MS, accounting for more than 99% of the total volatile fraction. Thymol and carvacrol were the main compounds, ranging from 10.0 + 0.9 to 43.8 + 3.8% and 4.5 + 0.4 to 39.1 + 3.8%, respectively, of the total. o-Cymene, γterpinene, and linalool were the most representative compounds in all the EOs. Keywords: Thymus serrulatus Hochst. ex Benth., Lamiaceae, essential oil, thymol, carvacrol, statistics. The genus Thymus (Lamiaceae) includes about 350 species worldwide and is widely distributed in temperate areas [1]. T. vulgaris contains 0.8-2.6% (usually ca. 1%) volatile oil, consisting of highly variable amounts of phenols, monoterpene hydrocarbons, and alcohols. There are several reports on the chemical composition of thyme oils; many indicate either thymol or carvacrol as the major compounds in the oils [1-23]. The leaves of T. vulgaris are used as an herb in food preparations, while the essential oil from the leaves is used in the alimentary, cosmetic and pharmaceutical industries. Thyme oil is used as an antispasmodic, carminative, antiseptic, anthelmintic, expectorant, antimicrobial (broad-spectrum antibacterial, antifungal and antiviral activity), antirheumatic, antioxidative, and natural food preservative [10,2444]. The strong antimicrobial activity of thyme oil is ascribed mainly to the high content of phenolic constituents, such as thymol and carvacrol [24]. The essential oil of T. vulgaris has potent repellent activity against Culex pipiens pallens [27]. Two species, T. schimperi Ronninger and T. serrulatus Hochst. ex Benth., are indigenous to Ethiopia [16], while T. vulgaris has been recently introduced. Chemical polymorphism concerning the essential oils of the genus Thymus is a widespread phenomenon. For example, the two Finnish species, T. serpyllum var. serpyllum and T. serpyllum var. tanaenis, turned out to form 4 different chemotypes each, with hedycaryol, germacra-1(10),5-dien-4-ol, germacra1(10),4-dien-6-ol, linalool, and linalyl acetate as type-characterizing compounds. T. schimperi and T. serrulatus belong to the thymol-carvacrol chemotypes [16]. The leaves of T. serrulatus are used in Ethiopia as spices to flavor food, as well as for medicines. People in Bale harvest T. serrulatus for making a tea [16]. The aim of this study was to evaluate the essential oils of wild plants of T. serrulatus, collected from seven different areas of the Ethiopian plateaus and to acquire information on the thyme population by statistical methods. Table 1 reports the mean chemical analysis of five populations and includes all compounds found in the EOs from leaves and flowers. Fifty-three compounds were identified, which accounted for more than 99% of the total 2070 Natural Product Communications Vol. 3 (12) 2008 volatile fraction from plants of T. serrulatus. Either thymol or carvacrol were the main compounds, with average amounts ranging from 10.0 + 0.9 to 43.8 + 3.8% and from 4.5 + 0.4 to 39.1 + 3.8%, respectively. O-cymene, γ-terpinene, and linalool were the most representative compounds in all the EOs. Cluster analysis of the database, including a compound selection, put in evidence the presence of four “natural” groups: area 3 is joined with area 6 and 7, area 2 with area 4, while area 5 and area 1 remain ungrouped (Figure 1). The analysis shows that areas 3-6-7 join at a distance cluster (ds) = 38.88, while areas 2 and 4 join at a ds = 36.66. The first clusters were formed at a distance cluster (da) = 1.03. Five major compounds listed in Table 2, which accounted for more than 75% of the essential oils, were selected to discuss the chemical variability due to areas of origin. Each area could be characterized by the chemical composition trend of the five major compounds. In Z1, o-cymene and carvacrol have the same percent value, in Z2 carvacrol is comparable to linalool and achieves its minimum value, in Z3 linalool achieves the minimum value, in Z4 carvacrol is comparable with γ-terpinene, in Z5 carvacrol is comparable to o-cymene, in Z6 linalool and γ-terpinene are comparable, and in Z7 no one compound is comparable to another. These remarks were supported by the ANOVA applied to a database in which the cases were the values of the five compounds, while the variables were the areas (Z1-Z7). According to many authors [1-16], there are two chemotypes of T. serrulatus. The first one comprises the oils with a high thymol concentration and was in accord with the plants collected in Z2, Z4, and Z5 areas. The second one, with a high carvacrol content, corresponds to the plants collected in Z1, Z6, and Z7 areas. There is a question that might be of interest for these data: do the measurements of the main compounds discriminate between the two assumed groups of oils and can they be used to produce a useful rule for classifying other oils, such as those from Z3 areas or other oils that might become available? The Fisher’s linear discriminant function analysis (FLDFA) works with data that are already classified into groups to derive rules for classifying new (and as yet unclassified) individuals on the basis of their observed variable values. The within-group covariance matrices suggest that the sample values differ to some extent, but according to Box’s test for Tirillini et al. Z5 Z1 Z2 Z4 Z3 Z6 Z7 Figure 1: Dendrogram using Average Linkage (between groups) equality of covariances, these differences are not statistically significant. The canonical correlation value is 0.988 so that 97.6% of the variance in the discriminant function scores can be explained by group differences. In the Wilk’s Lambda test, the lambda coefficient was 2.5%, and is the proportion of the total variance in the discriminant scores not explained by differences among the groups. The Fisher’s linear discriminant function is: z = -4.92 x meas1 – 20.84 x meas2 – 15.39 x meas3 + 79.21 x meas4 –74.99 x meas5. The threshold against which an oil discriminant score is evaluated is –5.00. Thus, new oils with discriminant scores above –5.00 would be assigned to the thymol type; otherwise, they would be classified as carvacrol type. Following the derived classification rule, the oils from Z3 areas would be assigned to the carvacrol type. In conclusion, if a representative lot of a plant population was hydrodistilled, a statistical approach may be useful for population inference. The evaluation of essential oils from wild plants living in different geographic areas could be helpful in the chemotype classification; the analyzed oils might be divided into two types according to the relative amounts of thymol or carvacrol. The “natural” group of oil did not correspond to the geographical areas as arbitrary chosen, but it might be possible redefine the areas in accordance with the cluster analysis. When the thyme population is distributed in an heterogeneous way across a large area, it might be possible to correlate the percent oil composition to the living sample area. Essential oils from Thymus serrulatus Natural Product Communications Vol. 3 (12) 2008 2071 Table 1: Chemical composition (area percent + SD) of T. serrulatusa. . Compounds b Methyl-2-methyl-butyrate α-Thujene RI c 778 931 Z1 % +SD 0.07 2.3 Z2 % + SD Z3 % + SD Z4 % + SD Z5 % + SD Z6 % + SD Z7 % 0.01 0.2 0.1 2.8 0.02 0.26 0.1 3.1 0.01 0.3 0.1 1.7 0.01 0.2 Tr 0.8 0.07 Tr 1.7 0.1 + SD 0.06 2.0 0.01 0.2 α-Pinene 939 1.0 0.08 0.4 0.03 0.4 0.03 0.2 0.02 0.09 0.01 0.3 0.02 0.4 0.04 Camphene 954 0.07 0.01 Tr - Tr - Tr - Tr - Tr - 0.04 0.01 2,4(10)-Thujadien 960 0.07 0.01 0.08 0.01 Tr - 0.04 0.01 Tr - Tr - Tr - Sabinene 975 0.06 0.01 0.2 0.02 0.2 0.02 0.06 0.01 Tr - 0.1 0.01 0.2 0.01 β-Pinene 980 0.2 0.02 0.09 0.01 0.09 0.01 0.06 0.01 Tr - 0.08 0.01 0.1 0.01 1-octen-3-ol 982 0.1 0.01 0.9 0.06 0.7 0.07 0.5 0.05 0.09 0.01 0.1 0.01 0.2 0.02 3-Octanone 985 3.6 0.3 1.6 0.13 1.8 0.2 1.0 0.09 0.4 0.04 2.7 0.2 3.4 0.3 Myrcene 991 1.7 0.2 1.7 0.15 1.3 0.09 1.0 0.1 0.6 0.05 0.9 0.09 0.8 0.08 3-Octanol 992 1.2 0.1 0.7 0.05 0.6 0.06 0.4 0.04 0.1 0.01 0.8 0.07 0.9 0.08 α-Phellandrene 1003 0.4 0.04 0.4 0.04 0.3 0.03 0.3 0.02 Tr - 0.1 0.01 0.2 0.02 α-Terpinene 1016 2.6 0.3 3.7 0.27 3.0 0.3 2.4 0.2 1.6 0.1 1.7 0.1 1.9 0.2 o-Cymene 1025 22.5 1.9 28.4 2.36 37.2 2.7 18.7 1.5 16.4 1.5 27.1 2.1 26.0 2.5 Limonene 1031 0.4 0.04 0.5 0.05 0.5 0.05 0.3 0.03 Tr - 0.3 0.02 0.3 0.02 β-Phellandrene 1032 0.3 0.03 0.3 0.03 0.3 0.03 0.2 0.02 Tr - 0.2 0.02 0.3 0.02 1,8-Cineole (Z)-β-Ocimene 1033 1036 0.4 0.4 0.04 0.02 0.1 0.2 0.01 0.02 Tr 0.1 0.02 0.1 0.1 0.01 0.01 Tr Tr - Tr 0.2 0.02 0.06 0.2 0.01 0.02 (E)-β-Ocimene 1051 0.1 0.02 0.1 0.01 Tr - 0.09 0.01 Tr - Tr - Tr - γ-Terpinene 1061 15.6 1.3 22.8 2.06 8.0 0.7 13.7 1.3 12.0 1.1 4.1 0.3 3.1 0.2 cis-Sabinene hydrate 1069 0.9 0.07 1.0 0.1 1.4 0.1 0.8 0.08 0.8 0.06 1.5 0.1 1.8 0.2 cis-Linalool oxide 1087 0.07 0.01 Tr - Tr - Tr - Tr - Tr - 0.04 0.01 Terpinolene 1089 0.1 0.01 0.2 0.02 Tr - 0.1 0.01 Tr - Tr - 0.03 0.01 Linalool 1098 6.2 0.6 5.0 0.5 1.7 0.1 3.3 0.3 3.3 0.2 3.6 0.3 4.8 0.4 (3Z)-Hexenyl isobutanoate 1147 0.2 0.02 Tr - Tr - 0.2 0.02 Tr - Tr - Tr - Borneol 1170 Tr - Tr - Tr - Tr - Tr - Tr - 0.03 0.01 Terpinen-4-ol 1177 1.04 0.08 0.7 0.05 0.6 0.05 0.2 0.02 0.4 0.03 0.2 0.02 0.1 0.01 p-Cymen-8-ol 1183 Tr - Tr - Tr - 0.06 0.01 Tr - Tr - Tr - α-Terpineol 1189 0.7 0.07 0.5 0.05 0.6 0.05 0.6 0.05 0.4 0.02 0.2 0.02 0.4 0.04 cis-Dihydrocarvone 1193 0.1 0.01 Tr - Tr - 0.04 0.01 Tr - Tr - 0.07 0.01 Thymol. methyl ether 1235 0.2 0.02 0.2 0.02 0.2 0.02 0.1 0.02 Tr - 0.3 0.02 0.2 0.02 Linalyl acetate 1257 0.06 0.01 4.0 0.36 Tr - 2.0 0.1 2.3 0.2 Tr - Tr - Thymol 1290 10.1 0.8 16.3 1.3 14.9 1.4 34.7 3.1 43.8 3.8 11.5 1.0 10.0 0.9 Carvacrol 1299 20.6 1.8 4.5 0.37 19.1 1.6 13.9 1.0 15.0 1.2 39.1 3.8 38.0 3.3 Thymol acetate 1352 0.3 0.03 0.3 0.03 Tr - 0.4 0.04 0. 0.01 Tr - 0.06 0.01 Carvacrol acetate 1373 2.3 0.2 Tr - Tr - 0.1 0.01 Tr - 0.1 0.02 0.2 0.02 β-Bourbonene 1388 0.05 0.01 Tr - Tr - Tr - Tr - Tr - Tr - α-Gurjunene 1410 0.07 0.01 Tr - Tr - Tr - Tr - Tr - Tr - (E)-Caryophyllene 1419 1.3 0.1 1.3 0.11 1.7 0.1 1.2 0.1 1.3 0.1 1.2 0.1 1.5 0.1 0.03 α-trans-Bergamotene 1435 Tr - 0.1 0.01 0.3 0.02 0.1 0.01 Tr - 0.2 0.02 0.3 Aromadendrene 1441 0.9 0.07 Tr - Tr - Tr - Tr - Tr - Tr - α-Humulene 1455 Tr - Tr - 0.1 0.01 Tr - Tr - Tr - 0.08 0.01 allo-Aromadendrene 1460 0.1 0.01 Tr - Tr - Tr - Tr - Tr - 0.05 0.01 γ-Muurolene 1480 0.06 0.01 Tr - Tr - Tr - Tr - 0.2 0.02 0.0 0.01 Germacrene D 1485 0.07 0.01 0.1 0.01 0.3 0.03 0.04 0.01 Tr - Tr - 0.2 0.02 Viridiflorene 1497 0.07 0.01 Tr - Tr - Tr - Tr - Tr - Tr - Bicyclogermacrene 1500 Tr - Tr - 0.1 0.01 0.06 0.01 Tr - 0.1 0.01 0.2 0.02 0.02 β-Bisabolene 1506 0.1 0.01 Tr - 0.08 0.01 0.1 0.01 Tr - 0.1 0.02 0.2 γ-Cadinene 1512 0.09 0.01 Tr - Tr - 0.08 0.01 Tr - Tr - Tr - Δ-amorphene 1513 0.1 0.01 Tr - 0.1 0.01 0.1 0.01 Tr - Tr - 0.04 0.01 β-Sesquiphellandrene 1525 0.4 0.04 0.06 0.01 0.3 0.03 0.3 0.02 Tr - 0.3 0.03 0.4 0.04 Spathulenol 1577 Tr - Tr - Tr - Tr - Tr - Tr - 0.06 0.01 Caryophyllene oxide 1585 0.1 0.01 Tr - Tr - 0.1 0.01 Tr - 0.2 0.02 0.2 0.02 Total a 99.96 99.68 99.59 99.99 99.79 99.52 99.48 essential oils of plants collected from the Z1-Z7 areas. b Compounds were listed in order of their elution from a DB-5MS column. c RI, retention indices as determined on DB-5MS column using homologous series of n-alkanes. Tr= trace (< 0.01%). 2072 Natural Product Communications Vol. 3 (12) 2008 Table 2: Chemical variability of the major compounds of T. serrulatus essential oil from Z1-Z7 areas. Values within a row for each compound having different letters are significantly different from each other using Tukey’s LSD test (P<0.05). Compounds Z1 Z2 Z3 Z4 Z5 Z6 Z7 o-Cymene γ-Terpinene Linalool thymol Carvacrol 3.11c 2.75 e 1.80 d 2.30 a 3.02 c 3.30 d 3.07 f 1.60 c 2.80 c 1.48 a 3.59 e 2.01 c 0.51 a 2.73 c 2.93 c 2.96 b 2.56 d 1.21 b 3.50 d 2.63 b 2.79 a 2.49 d 1.16 b 3.75 e 2.65 b 3.26 cd 1.45 b 1.22 b 2.46 b 3.60 d 3.24 cd 1.1 a 1.56 c 2.26 a 3.63 d Experimental Plant materials: Leaves and flowers of T. serrulatus growing in different area of Ethiopia were collected in May-June 2007 and dried at ambient temperature. A large number of plants (>5 kg) were randomly collected over the same area. The plants were collected in seven areas (Z1-Z7). Voucher specimens were deposited in the Herbarium of the CAMS – Univ. of Perugia (IPO-E1-05-07). Extraction of oil: The plants were subjected to hydrodistillation using a Clevenger-type apparatus for 3 h yielding 0.8 + 0.1% (mean value) of a yellowish oil. The oil was dried over anhydrous sodium sulfate and stored in sealed vials under refrigeration prior to analysis. GC and GC-MS analysis: The GC analyses were carried out using an Agilent 6890N instrument equipped with a FID and an HP-InnoWax capillary column (30 m x 0.25 mm, film thickness 0.17 μm), working from 60°C (3 min) to 210°C (15 min) at 4°C/min or an DB-5MS capillary column (30 m x 0.25 mm, film thickness 0.25 μm) working from 60°C (3 min) to 300°C (15 min) at 4°C/min; injector and detector temperatures, 250°C; carrier gas, helium (1 mL/min); split ratio, 1 : 10. GC-MS analyses were carried out using an Agilent 5975 GC-MS system operating in the EI mode at 70 eV, using the two above mentioned columns. The operating conditions were analogous to those reported in the GC analyses section. Injector and transfer line temperatures were 220°C and 280°C, respectively. Helium was used as the carrier gas, flow rate 1 mL/min. Split ratio, 1 : 10. Identification of the components: The identification of the components was made by matching their Tirillini et al. spectra with those from mass spectral libraries and the identity of each component was confirmed by comparing their retention indices, for both columns, relative to the C6-C22 n-alkanes with those from the literature. When reported, co-elution gas chromatography with reference compounds was used for an additional confirmation of the compound identity. The percentage composition of the essential oil was obtained by the normalization method from the GC peak areas, without using correction factors. Experimental design: The plants collected in each area were hydrodistilled separately and the ghost effect was minimized. The handling data was the percent concentration of the identified compounds. If we suppose that hydrodistillation of 7 batches of the same lot gives for each compound values +10% across the mean, from each percent composition of the 7 hydrodistilled oils, we might obtain 7 parent percent oil compositions. A 49% composition (7 oils for 7 areas (Z1-Z7)) for 53 oil compounds were the basic data file. Data file handling: Many problems arose from this type of database in a statistical analysis. The presence of compounds above 0.01% (tr) required a valuation: the addition to the database of 0.01, gave a statistical improvement and removed the null value. Compounds scarcely present in the database were removed. The independence of the data were checked using a linear or logaritmic coefficient of correlation between each pair of compounds: one of the two compounds with R >0.98 was removed (between limonene and β-phellandrene there was a high logarithmic correlation and β-phellandrene was removed). The great data dishomogeneity suggests the use of the Box-Cox method for the choice of most useful data transformation. The log-likelihood function suggests the use of natural logarithmic data transformation. According to Kolmogorov-Smirnov tests, the database in each group follows a normal distribution and according to the Levene test the variance of the database is the same in the groups (homoschedasticity assumption). Acknowledgments – We are grateful to IPO (Increasing People Opportunities) for providing us with the essential oils of T. serrulatus. References [1] Demissew S. (1993) The genus Thymus (Labiatae) in Ethiopia. Opera Botanica, 121, 57-60. Essential oils from Thymus serrulatus Natural Product Communications Vol. 3 (12) 2008 2073 [2] Arrebola ML, Navarro MC, Jimenez J, Ocana FA. (1994) Yield and composition of the essential oil of Thymus serpylloides subsp. serpylloides. Phytochemistry, 36, 67-72. [3] Asllani U. (1973) Albanian thyme varieties and their essential oils. Buletin i Shkencave Natyrore, 27, 111-129. [4] Baser KHC, Kirimer N, Ermin N, Ozek T, Tumen G. (1996) Composition of essential oils from three varieties of Thymus praecox Opiz growing in Turkey. Journal of Essential Oil Research, 8, 319-321. [5] Benjilali B, Hammoumi M, M'Hamedi A, Richard H. (1987) Essential oil composition of different Moroccan thyme varieties. 2 Principal component analysis. Sciences des Aliments, 7, 275-299. [6] Dagne E, Hailu S, Bisrat D, Worku T. (1998) Constituents of the essential oil of Thymus schimperi. The Bulletin of the Chemical Society of Ethiopia, 12, 79-82. [7] Delpit B, Lamy J, Rolland F, Chalchat JC, Garry RP. (1996) Common thyme (Thymus vulgaris). A 4-thujanol chemotype cloning and chemical study. Rivista Italiana EPPOS, 7(Spec. Num), 409-416. [8] Fan M, Chen J. (2002) Variations of essential oil and compositions of thyme collected at different time in Northwestern China. Xibei Zhiwu Xuebao, 22, 1451-1456. [9] Kasumov FY, Gadzhieva TG. (1980) Components of Thymus kotshyanus. Khimiya Prirodnykh Soedinenii, 5, 728-732. [10] Jimenez J, Navarro MC, Montilla MP, Martin A, Martinez A. (1993) Thymus zygis oil: its effects on carbon tetrachloride-induced hepatotoxicity and free radical scavenger activity. Journal of Essential Oil Research, 5, 153-158. [11] Kasumov FY, Gavrenkova SI. (1985) Thymus transcaucasicus Ronn - promising essential oil containing plant of Azerbaijan flora. Doklady - Akademiya Nauk Azerbaidzhanskoi SSR, 41, 56-59. [12] Kasumov FY, Farkhadova MT. (1986) The composition of Thymus karamarianucus essential oil. Khimiya Prirodnykh Soedinenii, 5, 642-643. [13] Kulevanova S, Ristic M, Stafilov T, Dorevski K, Ristov T. (1996) Essential oil analysis of some taxa of genera Thymus L. environment influences. Glasnikna Hemicarite i Tehnolozite na Makedonija, 15, 33-38. [14] Lamy J. (1983) Thyme breeding in the Drome Region [France]. Parfums, Cosmetiques, Aromes, 51, 73-74. [15] (a) Miquel JD, Richard HMJ, Sandret FG. (1976) Volatile constituents of Moroccan thyme oil. Journal of Agricultural and Food Chemistry, 24, 833-835; (b) Nigisti A, Storesund HJ, Scattebol L, Tonnesen F, Aasen AJ. (1999) Volatile oil constituents of two Thymus species from Ethiopia. Flavour and Fragrance Journal, 15, 123-125. [16] (a) Miguel MG, Costa LA, Figueiredo AC, Barroso JG, Pedro LG (2007) Assessment of the antioxidant activity of Thymus albicans, T. mastichina, T. camphoratus and T. carnosus essential oils by TBRAS and micellar model systems. Natural Product Communications, 2, 399-406; (b) Stoilova I, Bail S, Buchbauer G, Krastanov A, Stoyanova A, Schmidt E, Jirovetz L (2008) Natural Product Communications, 3, 1047-1050. [17] Park BS, Choi WS, Kim JH, Kim KH, Lee SE. (2005) Monoterpenes from thyme (Thymus vulgaris) as potential mosquito repellents. Journal of the American Mosquito Control Association, 21, 80-83. [18] Pino JA. (1999) Volatile components in spices. XV. Thyme. Alimentaria (Madrid), 301, 107-109. [19] Passet J. (1979) Chemical variability within thyme, its manifestations and its significance. Parfums, Cosmetiques, Aromes, 28, 39-42. [20] Raghavan B, Abraham KO, Koller WD. (1995) Flavor quality of fresh and dried Indian thyme (Thymus vulgaris L.). PAFAI Journal, 17, 9-14. [21] Russell GF, Olson KV. (1972) Volatile constituents of oil of thyme. Journal of Food Science, 37, 405-407. [22] Saez F. (1995) Essential oil variability of Thymus hyemalis growing wild in southeastern Spain. Biochemical Systematics and Ecology, 23, 431-438. [23] Stoeva T, Dobos A, Mathe I, Bosseva Y. (2001) Productivity and composition of essential oils of Bulgarian thyme varieties. Rasteniev"dni-Nauki (Bulgaria). Plant Science, 38, 359-361. [24] Lattaoui N, Tantaoui-Elaraki A. (1994) Comparative kinetics of microbial destruction by essential oils of Thymus brousskonettii, Thymus zygist and Thymus satureioides. Journal of Essential Oil Research, 6, 165-171. [25] Bagamboula CF, Uyttendaele M, Debevere J. (2004) Inhibitory effect of thyme and basil essential oils, carvacrol, thymol, estragol, linalool and p-cymene towards Shigella sonnei and S. flexneri. Food Microbiology, 21, 33-42. [26] Braga PC, Dal Sasso M, Culici M, Galastri L, Marceca MT, Guffanti EE. (2006) Antioxidant potential of thymol determined by chemiluminescence inhibition in human neutrophils and cell-free systems. Pharmacology, 76, 61-68. [27] Choi WS, Park BS, Ku SK, Lee SE. (2002) Repellent activities of essential oils and monoterpenes against Culex pipiens pallens. Journal of the American Mosquito Control Association, 18, 348-351. [28] Choi WS, Park BS, Lee YH, Jang DY, Yoon HY, Lee S-E. (2006) Fumigant toxicities of essential oils and monoterpenes against Lycoriella mali adults. Crop Protection, 25, 398-401. [29] Dal Sasso M, Culici M, Braga PC, Guffanti EE, Mucci M. (2006) Thymol: inhibitory activity on Escherichia coli and Staphylococcus aureus adhesion to human vaginal cells. Journal of Essential Oil Research, 18, 130-136. 2074 Natural Product Communications Vol. 3 (12) 2008 Tirillini et al. [30] Santoro GF, Gracas Cardoso M, Guimaraes LG, Salgado AP, Menna-Barreto RF, Scares MJ. (2007) Effect of oregano (Origanum vulgare L.) and thyme (Thymus vulgaris L.) essential oils on Trypanosoma cruzi (Protozoa: Kinetoplastida) growth and ultrastructure. Parasitology Research, 100, 783-790. [31] Fan M, Chen J. (2001) Study on antimicrobial activity of extract from Thyme. Weishengwu Xuebao, 41, 499-504. [32] Ferrarese L, Uccello A, Zani F, Ghirardini A. (2006) Antimicrobial activity and phytocosmetic applications of the essential oil of Thymus vulgaris L. Cosmetic News, 29, 301-304. [33] Ibragimov GG, Vasil'ev OD. (1985) Antifungal activity of essential oils. Azerbaidzhanskii Meditsinskii Zhurnal, 62, 44-48. [34] Kasumov FY, Ismailov NM. (1976) Biochemical study of the essential oils of trans-Caucasian thyme and development of its useful properties. Seriya Biologicheskikh Nauk, 6, 26-31. [35] Manou I, Bouillard L, Devleeschouwer MJ, Barel AO. (1998) Evaluation of the preservative properties of Thymus vulgaris essential oil in topically applied formulations under a challenged test. Journal of Applied Microbiology, 84, 368-376. [36] Motiejunaite O, Kalediene L. (2003) Antimicrobial activity of Lamiaceae plant essential oils on Aspergillus niger growth. Bulletin of the Polish Academy of Sciences: Biological Sciences, 51, 237-242. [37] Pank F, Eichholz E, Ennet D. (1982) Chemical weed control in the cropping of medicinal plants. Part 4. Thyme (Thymus vulgaris L.). Pharmazie, 37, 795-797. [38] Segvic KM, Kosalec I, Mastelic J, Pieckova E, Pepeljnak S. (2007) Antifungal activity of thyme (Thymus vulgaris L.) essential oil and thymol against moulds from damp dwellings. Letters in Applied Microbiology, 44, 36-42. [39] Singh A, Singh RK, Bhunia AK, Singh N. (2003) Efficacy of plant essential oils as antimicrobial agents against Listeria monocytogenes in hotdogs. Lebensmittel-Wissenschaft und -Technologie, 36, 787-794. [40] Stahl-Biskup E. (2002) Thyme as a herbal drug - pharmacopeias and other product characteristics. Medicinal and Aromatic PlantsIndustrial Profiles, 24 (Thyme), 293-316. [41] Tanovic B, Milijasevic S, Todorovic B, Potocnik I, Rekanovic E. (2005) Toxicity of essential oils to Botrytis cinerea Pers. in vitro. Pesticidi i Fitomedicina, 20, 109-114. [42] Uyttendaele M, Neyts K, Vanderswalmen H, Notebaert E, Debevere J. (2004) Control of Aeromonas on minimally processed vegetables by decontamination with lactic acid, chlorinated water, or thyme essential oil solution. International Journal of Food Microbiology, 90, 263-271. [43] Wagner A, Strudzinska A, Struszczyk H. (2003) Effect of some natural compounds on Alternaria alternata Keiss. and Botrytis cinerea Pers. Bulletin of the Polish Academy of Sciences: Biological Sciences, 51, 287-290. [44] Youdim KA, Deans SG, Finlayson HJ. (2002) The antioxidant properties of thyme (Thymus zygis L.) essential oil: an inhibitor of lipid peroxidation and a free radical scavenger. Journal of Essential Oil Research, 14, 210-215. NPC Natural Product Communications GC MS Analysis of the Volatile Constituents of Essential Oil and Aromatic Waters of Artemisia annua L. at Different Developmental Stages 2008 Vol. 3 No. 12 2075 - 2078 Anna Rita Biliaa,*, Guido Flaminib, Fabrizio Morgennic, Benedetta Isacchia and Franco FrancescoVincieria a Department of Pharmaceutical Sciences, via Ugo Schiff,6, University of Florence, 50019 Sesto Fiorentino (FI), Italy b Department of Bioorganic Chemistry and BioPharmacy, Università di Pisa, Via Bonanno 33, 56126 Pisa, Italy c Officina Profumo Farmaceutica di Santa Maria Novella, SRL, via Reginaldo Giuliani 141, 50141 Firenze, Italy ar.bilia@unifi.it Received: July 22nd, 2008; Accepted: October 31st, 2008 Artemisia annua L. (Asteraceae) still represents the only source of artemisinin, considered as one of the most important drugs for the treatment of malaria and which, more recently, has been shown to be effective against numerous types of tumors. The foliage and inflorescence of A. annua also yield an essential oil upon hydrodistillation. This oil has been evaluated at different development stages (pre-flowering and flowering) by GC/MS. The volatile oil from plants at full blooming showed numerous constituents, with germacrene D (21.2%), camphor (17.6%), β-farnesene (10.2%), β-caryophyllene (9%), and bicyclogermacrene (4.2%) among the main ones. Aromatic waters, after extraction with n-hexane, showed the presence, among others, of camphor (27.7%), 1,8-cineole (14%), artemisia ketone (10.1%), α-terpineol (6.1%), trans-pinocarveol (5.4%), and artemisia alcohol (2%). From plants at the pre-flowering stage, aromatic waters were obtained with camphor (30.7%), 1,8-cineole (12.8%), artemisia alcohol (11.4%), artemisia ketone (9.5%), alpha-terpineol (5.8%), and trans-pinocarveol (3.0%) as the main constituents. The qualitative and quantitative profiles of the two aromatic waters were similar. These results permitted the conclusion to be made that A. annua could be harvested a long time before the onset of flowering to obtain higher yields of artemisinin or could be allowed to attain maturity to obtain valuable yields of volatiles. Keywords: Artemisia annua, Asteraceae, volatile constituents, essential oil, aromatic waters, pre-flowering and blossom time, GC-MS analysis. Artemisia annua L.(family Asteraceae), indigenous to south east Asia, is an annual herb/shrub, which has become naturalized or is cultivated as a horticultural or medicinal plant in many parts of Asia, Africa, Europe, America and Australia. It has been used in Traditional Chinese Medicine for many centuries for the treatment of fever and malaria due to the presence in the leaves and capitula of a unique sesquiterpene endoperoxide called artemisinin. Nowadays, A. annua still represents the only source of artemisinin [1,2], considered to be one of the most important drugs for the treatment of malaria and, more recently, it has been shown to be effective also against numerous types of tumors, including breast cancer, human leukemia, colon, and small-cell lung carcinomas [3]. It is well known that natural populations and genetic resources of A. annua from different areas have considerable variability in the accumulation of artemisinin in the leaves and the capitula of the plant, with an artemisinin content ranging from 0.001 to 0.8% [1]. The foliage and inflorescence of A. annua plants also yield an essential oil upon hydrodistillation, which could represent another potential commercially valuable product [4,5]. In this current study the volatile constituents of both the essential oil and aromatic waters obtained by hydrodistillation of 2076 Natural Product Communications Vol. 3 (12) 2008 Bilia et al. Table 1: Identified constituents of the investigated samples. l.r.i. (A) Constituents (E)-3-Hexen-1-ol α-Pinene Camphene Sabinene β-Pinene Myrcene 2,3-Dehydro-1,8cineole Yomogi alcohol (E)-3-Hexen-1-yl acetate α-Terpinene p-Cymene Santolina alcohol 1,8-Cineole γ-Terpinene Artemisia ketone cis-Sabinene hydrate Artemisia alcohol Linalool trans-Sabinene hydrate Dehydrosabinaketone α-Campholenal cis-p-Menth-2-en-1-ol trans-Pinocarveol trans-p-Menth-2-en-1ol Camphor Camphene hydrate β-Pinene oxide Sabinaketone Pinocarvone δ-Terpineol Borneol Terpinen-4-ol p-Cymen-8-ol α-Terpineol Myrtenol Verbenone (E,E)-2,4-Nonadienal trans-Carveol Thymyl methyl oxide cis-Carveol Carvone cis-Chrysanthenyl acetate Lavandulyl acetate Isobornyl acetate Thymol trans-Carvyl acetate Eugenol cis-Carvyl acetate α-Copaene β-cubebene Benzyl valerate (E)-β-Caryophyllene (E)-β-Farnesene α-Humulene 853 941 956 978 982 992 994 998 Table 1 (Contd.) n-Hexane extract of aromatic waters. Plants at preflowering n-Hexane extract of aromatic waters. Plants at full blooming Volatile oil -full blooming 1373 1027 1071 1116 1108 1145 0.2 tr 0.1 tr 0.1 nd 0.1 0.4 0.2 0.2 0.1 0.5 tr 0.7 1.5 0.2 0.5 0.1 - 0.1 2.5 nd 1.2 tr nd l.r.i. (B) 1401 - 1008 1020 1029 1033 1035 1064 1065 1071 1084 1101 1103 1123 1127 1129 1141 1183 1245 1413 1208 1249 1355 1462 1504 1557 1458 1641 nd nd tr 0.4 12.8 nd 9.5 0.7 11.4 1.1 1.0 0.8 tr 0.2 3.0 nd nd 0.1 0.2 14.0 nd 10.1 0.2 2.0 3.9 0.6 0.9 tr 0.2 5.4 0.1 0.1 1.7 nd 1.4 0.3 nd 0.2 nd 0.2 0.2 nd tr nd nd 1142 1148 1153 1159 1161 1166 1169 1171 1180 1186 1192 1197 1207 1220 1222 1233 1234 1245 1560 1522 1611 1549 1676 1796 1607 1835 1688 1791 1715 1705 1874 1593 1845 1742 nd 30.7 nd 0.7 0.2 3.2 0.9 3.9 2.4 0.2 5.8 0.1 0.2 0.8 0.8 0.1 0.2 0.3 nd 27.7 nd 2.5 0.4 3.8 1.0 1.8 3.4 0.4 6.1 0.2 tr 0.6 0.5 0.7 0.3 0.3 1.3 17.6 0.1 nd nd 0.3 nd 0.9 0.6 nd 0.5 nd nd nd 0.3 nd tr nd 1263 1286 1286 1293 1339 1358 1363 1377 1389 1391 1419 1456 1457 1596 1582 2186 1756 2173 1793 1475 1545 1604 1660 1666 nd nd 0.7 nd nd 1.1 nd nd nd 0.3 tr nd tr nd nd 0.2 nd nd 1.1 nd 0.2 nd 0.3 0.6 0.3 tr 0.4 0.9 nd 1.5 1.0 tr 0.4 0.9 0.7 nd 9.0 10.2 0.1 β-Chamigrene Germacrene D β-Selinene Bicyclogermacrene δ-Cadinene trans-Nerolidol Spathulenol Caryophyllene oxide Globulol epi-Cedrol β-Acorenol Cubenol T-Muurolol Kongol α-Cadinol Elemol acetate (Z)-α-Santalol Total identified 1476 1483 1489 1496 1524 1563 1578 1583 1586 1599 1640 1643 1645 1662 1655 1663 1680 1757 1709 1713 1736 1731 1996 2136 2070 2054 2139 2045 2150 2236 2187 2310 tr tr tr nd nd nd 0.5 0.3 nd nd 0.2 nd nd nd nd nd nd 0.1 0.4 0.3 nd nd nd 0.7 0.4 nd nd nd nd nd 0.1 nd nd nd 1.4 21.2 0.8 4.2 0.5 0.4 1.3 0.6 0.2 0.5 nd 2.3 0.7 0.6 0.5 0.5 1.3 97.5 95.0 91.2 (A): linear retention index (l.r.t) obtained with a phenylmethylsilicone column (apolar) (B): linear retention index (l.r.t) obtained with a PEG column (polar) A annua collected at different developmental stages (pre-flowering and full blooming) have been evaluated by GC/MS. The aim of this study was the evaluation of the essential oil and aromatic waters as ingredients of food, pharmaceutical and cosmetic products, depending on the composition of the volatile constituents. Fresh, wild plant materials were collected near Sesto Fiorentino (FI, Italy) at pre-flowering and full blooming in August and September 2007 and submitted to hydrodistillation. Only samples in full bloom gave essential oil, which was analysed by GC/MS. Aromatic waters obtained from both samples were also analysed after extraction with n-hexane. Seventy-two compounds were identified in the different samples, accounting for 91.2%-97.5% of the total compositions. Volatile oil from plants at full bloom showed numerous constituents, with germacrene D (21.2%), camphor (17.6%), β-farnesene (10.2%), βcaryophyllene (9%), and bicyclogermacrene (4.2%) among the main ones. Aromatic waters, after extraction with n-hexane, showed the presence, among others, of camphor (27.7%), 1,8-cineole (14%), artemisia ketone (10.1%), alpha-terpineol (6.1%), trans-pinocarveol (5.4%), and artemisia alcohol (2%). From plants at the pre-flowering stage, only aromatic waters were obtained with camphor (30.7%), 1,8-cineole (12.8%), artemisia alcohol (11.4%), artemisia ketone (9.5%), α-terpineol (5.8%), and trans-pinocarveol (3.0%) as their main GC-MS analysis of volatile constituents of Artemisia annua Natural Product Communications Vol. 3 (12) 2008 2077 constituents. Artemisinin was never detected in either the essential oil or aromatic waters. The qualitative and quantitative profiles of the two aromatic waters were similar. Constituents found in our sample of essential oil were very similar to those reported in the literature, namely camphor, germacrene D, and artemisia ketone. In the aromatic waters 1,8 cineole was also found and can represent a good source of volatiles, to be used for different applications, including cosmetic, alimentary and pharmaceutical ones. In the literature there are several studies reporting the GC analysis of the essential oil obtained from different parts of A. annua of different origins [4-20], but none concerning the analysis of aromatic waters. A great variability in the qualitative and quantitative composition has been evidenced and apart from ecological factors, plant part and development stage, a main source of variability was the wild plant material or selected cultivar. The majority of the studies have been performed on plant material from India and the main compounds identified in the essential oils from the aerial parts were camphor (0-44.4%), 1,8 cineole (1.7-28.6%), artemisia ketone (0-52.9%), 2,5-dihydro-3-methylfuran (0-68.5%), camphene (0-28.4%), and germacrene D (0-10.9%) [4, 6-12]. The principal constituents detected in the essential oil of plants from Iran, depending on the flowering stage, were camphor (14.3-48.0%), germacrene D (2.0-18.5%),1,8-cineole (5.8-17.3%), α-pinene (013.3%), β-selinene (0-10.4%), and β-caryophyllene (0-9.4%) [13-14]. Two studies from the USA took into account the two subspecies, the “glanded” and “glandless” ones. Artemisia ketone was found as the main component (0-35.6% in leaves, up to 56% in flowers). Other important volatiles were germacrene D (0-49.8%), 1,8-cineole (0-28.1%), α-pinene (0-26.7%), camphor (0-20.5%), and pinocarvone (0-15.8%) [5,15]. A few studies with European plant material were performed in France [16], the Netherlands [17] and Hungary [18-19]. All of them reported that artemisia ketone was the main constituent (11.9-63.9%), with the exception of plants grown from Vietnamese seeds, where it was completely absent. Other main constituents were artemisia alcohol (11.0-56.0%) and camphor (21.8%) in Hungarian plant material, germacrene D (2.0-18.5%) in plants collected in France and the Netherlands, and 1,8-cineole (5.114.7%) in plants from France. Another study on the essential oil of the fruits reported that sesquiterpenes were the most abundant chemicals, i.e. caryophyllene oxide (9.0%), caryophyllene (6.9%), (E)- β-farnesene (8.2%) and germacrene D (4.0%). However, this oil was only partially characterized, with only 52% of the components being identified [20]. Furthermore, the results obtained permitted us to conclude that A. annua could be harvested either a long time before onset of flowering to obtain higher yields of artemisinin or the crop could be allowed to attain maturity to obtain valuable yields of the essential oil. Experimental Plant material and samples: Artemisia annua subspecies “glanded” was identified by Dr Lia Pignotti of the Department of Vegetal Biology, University of Florence, where an authentic specimen is also deposited. About 1 kg fresh, wild plant material was collected near Sesto Fiorentino and immediately submitted to hydrodistillation. Only samples in full bloom gave essential oil (0.5%), which was analysed by GC/MS. Aromatic waters obtained from both samples were also analysed after extraction with n-hexane. GC-EIMS analysis: GC-EIMS analyses were performed with a Varian CP-3800 gaschromatograph equipped with a DB-5 capillary column (30 m x 0.25 mm; coating thickness 0.25 mm) and a Varian Saturn 2000 ion trap mass detector. Analytical conditions: injector and transfer line temperatures at 220 and 240°C respectively; oven temperature was programmed from 60°C to 240°C at 3°C/min; carrier gas helium at 1 mL/min; injection of 0.2 mL (10% n-hexane solution); split ratio 1:30. Identification of the constituents was based on comparison of the retention times with those of authentic samples, comparing their Linear Retention Indices relative to the series of n-hydrocarbons, and by computer matching against commercial [21] and home-made library mass spectra built up from pure substances and components of known essential oils and MS literature data [21-26]. Moreover, the molecular weights of all the identified substances were confirmed by GC-CIMS, using MeOH as CI ionizing gas. 2078 Natural Product Communications Vol. 3 (12) 2008 Bilia et al. References [1] [2] [3] [4] [5] [6] [7] [8] [9] [10] [11] [12] [13] [14] [15] [16] [17] [18] [19] [20] [21] [22] [23] [24] [25] [26] Ferreira JFS, Janick J. (1996) Distribution of artemisinin in Artemisia annua. In: Janick J. (Ed.), Progress in New Crops. AHAS Press, Arlington, VA, pp. 579-584. Gupta SK, Singh P, Bajpai P, Ram G, Singh D, Gupta MM, Jain DC, Khanuja SP, Kumar S. (2002) Morphogenetic variation for artemisinin and volatile oil in Artemisia annua. Industrial Crops and Products, 16, 217-224 Efferth T. (2007) Willmar Schwabe Award 2006: antiplasmodial and antitumor activity of artemisinin-from bench to bedside. Planta Medica, 73, 299-309. Ma C, Wang H, Lu X, Li H, Liu B, Xu G. (2007) Analysis of Artemisia annua L. volatile oil by comprehensive two-dimensional gas chromatography time-of-flight mass spectrometry. Journal of Chromatography A, 1150, 50-53. Tellez MR, Canel C, Rimando AM, Duke SO. (1999) Differential accumulation of isoprenoids in glanded and glandless Artemisia annua L. Phytochemistry, 52, 1035-1040. Goel D, Mallavarupu GR, Kumar S, Singh V, Ali M. (2007) Volatile metabolite compositions of the essential oil from aerial parts of ornamental and artemisinin rich cultivars of Artemisia annua. Journal of Essential Oil Research, 20, 147-152. Mukhtar HM, Ansari SH, Ali M, Mir SR, Abdin MZ, Singh P. (2007) GC-MS analysis of volatile oil of aerial parts of Artemisia annua Linn. Journal of Essential Oil Bearing Plants, 10, 168-171. De Magalhaes PM, Pereira B, Sartoratto A. (2004) Yields of antimalarial Artemisia annua L. species. Acta Horticoltura, 629, 421-424. Zhang Y, Zhang J, Yao J, Wang L, Huang A, Dong L. (2004) Studies on the chemical constituents of the essential oil of Artemisia annua L. from Xinjiang. Xibei Shifan Daxue Xuebao, Ziran Kexueban, 40, 67-69. Bagchi GD, Haider F, Dwivedi PD, Singh A, Naqvi AA. (2003) Essential oil constituents of Artemisia annua during different growth periods at monsoon conditions of subtropical north Indian plains. Journal of Essential Oil Research, 15, 248-250. Jain N, Srivastava SK, Aggarwal KK, Kumar S, Syamasundar KV. (2002) Essential oil composition of Artemisia annua L. 'Asha' from the plains of northern India. Journal of Essential Oil Research, 14, 305-307. Ali M, Siddiqui NA. (2000) Volatile oil constituents of Artemisia annua leaves. Journal of Medicinal & Aromatic Plant Sciences, 22, 568-571. Verdian-Rizi MR, Sadat-Ebrahimi E, Hadjiakhoondi A, Fazeli MR, Pirali HM. (2008) Chemical composition and antimicrobial activity of Artemisia annua L. essential oil from Iran. Faslnamah-i Giyahan-i Daruyi, 7, 58-62. Lari yazdi H, Khavarinejad RA, Roustaian AH. (2002) The composition of the essential oil of Artemisia annua L. growing wild in Iran. Faslnamah-i Giyahan-i Daruyi, 1, 41-48. Charles DJ, Cebert E, Simon JE. (1991) Characterization of the essential oil of Artemisia annua L. Journal of Essential Oil Research, 3, 33-39. Biougne J, Chalchat JC, Garry RP, Lamy J. (1993) Essential oil of Artemisia annua: Seasonal variations in chemical composition. Rivista Italiana EPPOS, 4, 622-629. Woerdenbag HJ, Bos R, Salomons MC, Hendriks H, Pras N, Malingre TM. (1993) Volatile constituents of Artemisia annua L. (Asteraceae). Flavour & Fragrance Journal, 8, 131-137. Hethelyi EB, Cseko IB, Grosz M; Mark G, Palinkas JJ. (1995) Chemical composition of the Artemisia annua essential oils from Hungary. Journal of Essential Oil Research, 7, 45-48. Hethelyi I, Ceseko I, Grosz M, Mark G, Palinkas JJ. (1994) Capillary gas chromatographic investigation of Artemisia annua essential oils. Olaj, Szappan, Kozmetika, 43, 103-106. Li R, Wang D, Liao H. (2007) Chemical constituents of essential oil from the fruits of Artemisia annua L. Zhongnan Yaoxue, 5, 230-232. Stenhagen E, Abrahamsson S, McLafferty FW. (1974) Registry of mass spectral data, J. Wiley & Sons, New York. Massada Y. (1976) Analysis of Essential Oils by Gas Chromatography and Mass Spectrometry, J. Wiley & Sons, New York. Jennings W, Shibamoto T. (1980) In Qualitative Analysis of Flavor and Fragrance Volatiles by Glass Capillary Chromatography, Academic Press, New York,. Davies NW. (1990) Gas chromatographic retention indices of monoterpenes and sesquiterpenes on methyl silicon and Carbowax 20M phases. Journal of Chromatography, 503, 1-24. Swigar AA, Silverstein RM. (1981) In Monoterpenes, Aldrich Chem. Comp., Milwaukee. Adams RP. (1995) Identification of Essential Oil Components by Gas Chromatography-Mass Spectroscopy, Allured Publ. Corp., Carol Stream, Illinois. NPC Natural Product Communications Do Non-Aromatic Labiatae Produce Essential Oil? The Case Study of Prasium majus L. 2008 Vol. 3 No. 12 2079 - 2084 Claudia Giuliania, Roberto Maria Pellegrinob, Bruno Tirillinic and Laura Maleci Binia,* a Department of Vegetal Biology, University of Florence, via La Pira 4, 50121 Florence, Italy b Department of Chemistry, Laboratory of Organic Chemistry, University of Perugia, via Elce di Sotto 8, 06123 Perugia, Italy c Institute of Botany, University of Urbino, via Bramante 28, 61029 Urbino, Italy maleci@unifi.it Received: July 7th, 2008; Accepted: November 19th, 2008 Prasium majus L. (Labiatae, Lamioideae) is considered a typical non-aromatic plant. In this work we examined the glandular trichomes present on leaves and inflorescences and the essential oils of plants growing along the Tuscan coast of Italy. The micromorphological study evidenced different types of trichomes responsible for the essential oil production. The essential oil compositions of leaves and flowers were analyzed by GC/MS and are here reported. Keywords: Prasium majus, Labiatae, Lamioideae, glandular trichomes, micromorphology, histochemistry, essential oil, GC/MS analysis. The Labiatae family contains about 200 genera of which 40 per cent possess aromatic properties. Owing to these features, the family has a wide economic importance and several species have been widely studied. Currently the family is divided into seven different subfamilies [1], among which the largest ones are Nepetoideae and Lamioideae. Aromatic plants are mostly included within Nepetoideae [2], while species belonging to Lamioideae are usually non-aromatic plants, showing either scarce or absent essential oil production [2]. The production of essential oil is associated with the presence of highly specialized secretory structures known as glandular trichomes. Two types of glandular trichomes are recognized: peltate and capitate [3,4]. Peltate hairs are considered the site of synthesis and storage of essential oil [5], while capitate hairs present a more complex hydrophilic secretion, in which mucopolysaccharides prevail [4,5]. Prasium majus L. (Lamioideae) is considered a typical non-aromatic plant, lacking peltate hairs and showing only capitate hairs [3,5]. This species is an evergreen perennial shrub with white to pale lilac flowers, and leaves with a distinct glossy green colour [6]; neither leaves nor flowers give off any scent. The plant grows in maquis, guarigue, among bushes or rocks, field boundaries and beside dry-stone walls, mainly facing the sea. This plant is widespread in the whole Mediterranean basin and in central and southern Portugal [7]; in Italy, in particular, it is found in southern regions and Sardinia and the northern limit of its distribution area is the Tuscan Archipelago [8]. Concerning the micromorphology, only the anatomy of nutlets has been investigated [9]. Few popular uses have been reported for this plant; it has been employed medicinally in Greece as a tranquilizer [10], and in Tunisia its leaves are used in popular medicine for their soothing properties [11]. The plant is also consumed as a raw food in Tunisia [11], in Crete it is stir-fried or used in traditional vegetable pies [12], and in Sicily (Palermo and Trapani provinces) its use is limited to rural communities [13]. Some non-volatile products have been isolated and identified in the plant [10,14,15]. Recently the 2080 Natural Product Communications Vol. 3 (12) 2008 Table 1: Distribution of the different types of glandular trichomes present on vegetative and reproductive organs of P. majus. Results: (-) absent; (±) scarce; (+) present; (++) abundant. Stem A B C + - Leaf/ Bract adax + - abax + - Calyx adax - abax + + ++ Corolla adax - abax ± + essential oil composition of plants from Greece was reported [16]. In this current study we examined specimens of P. majus collected from along the Tuscan coast (Italy), during the blooming period (April-June), in order to describe the glandular trichomes present on leaves and flowers and to determine their type of secretion, particularly of essential oil. The essential oils both from leaves and inflorescences were obtained by hydrodistillation and their compositions were determined by GC/MS analysis. Micromorphological analysis The glandular trichomes included peltate (type A) and capitate types (types B and C) (Figure 1). Stem and leaves bear only few (Table 1) short capitate trichomes (type B), widely diffused and described in the whole Labiatae family [4,5] (Figure 1). They consist of one basal epidermal cell, one stalk cell and a secretory head (25-30 μm in size) of four cells, with a small subcuticular space in which the secretion is temporarily stored. Histochemical staining (Table 2) indicated secretion of polysaccharides (Figure 2) and a small amount of essential oil. The secreting cells ultrastructure shows numerous Golgi bodies and an abundant rough endoplasmic reticulum (RER), involved in polysaccharidic secretion [17], and few electron dense plastids responsible for essential oil production [17] (Figure 3). On the inflorescences, especially on the abaxial surfaces of the calyx (Figure 1) and corolla, besides the described type B trichomes, other types of glandular hairs are observed: several type A and numerous type C (Table 1). Moreover, short uniseriate non-glandular hairs (type D) are present (Figure 1). Type A trichomes, unlike the typical peltate hairs, present an elongated basal epidermal cell which Giuliani et al. Table 2: Histochemical tests on the different types of glandular trichomes. Results: (-) negative; (±) scarce; (+) intense; (++) very intense. Staining Nile Red Fluoral Yellow NADI reagent Ruthenium Red Alcian Blue FeCl3 Target compounds Neutral lipids Total lipids Terpenes Polysaccharides Polysaccharides Polyphenols A + + ++ - B ± ± ± + + - C ++ ++ + + ± + forms a well developed stalk, so that these trichomes are raised on the epidermal surface (Figures 1 and 4). This uncommon feature was already observed in Salvia officinalis [18] and in several species of Stachys [19]. The neck cell, the broad glandular head (40-50 μm in size) of eight secreting cells and the large subcuticular space present the typical morphology quoted in the literature [4,5,17]. The secretion stored in the subcuticular space is composed of essential oil (Table 2), since it shows a strong positive reaction only to the Nadi reagent (Figure 4). The most striking ultrastructural features observed in the cytoplasm of the secreting cells are plastids with large starch granules (Figure 5), associated with smooth endoplasmic reticulum (SER). These cellular compartments are typically involved in essential oil production and transfer [17]. Type C long capitate trichomes (Table 1; Figure 1), observed also in several Stachys species [19], consist of one basal epidermal cell, a stalk of two-three cells and a multicellular head (40-60 μm in size) of sixeight cells. Each glandular cell is endowed with a small subcuticular space; the secretion is extruded to the outside from the subcuticular space and also from the whole external wall and flows along the stalk to the epidermis [19]. The secretion shows a complex composition (Table 2), since it contains polysaccharides, essential oil and polyphenols (Figures 6 and 7). In young trichomes, the glandular cells ultrastructure shows mitocondria, Golgi bodies, RER vesicles and multi-shaped plastids with starch granules (Figure 8). In mature trichomes, Golgi bodies and RER elements occur occasionally, while plastids, SER and lipidic droplets (Figure 9) can be observed. Therefore, these hairs present different types of secretion according to the different ages of the trichomes. Essential oils analysis Very small amounts of essential oils were obtained by hydrodistillation of the leaves and inflorescences; their compositions are reported in Table 3. Micromorphology and essential oil analysis of Prasium majus Natural Product Communications Vol. 3 (12) 2008 2081 Figure 1: Trichomes on the abaxial side of the calyx: A. peltate, B. short capitate, C. long capitate and D. simple uniseriate non-glandular trichomes. Bar = 100 μm. Figure 2: Histochemistry of type B trichome: Alcian Blue. Bar = 25 μm. Figure 3: Secreting cell cytoplasm of type B trichome. Bar = 1 μm. Figure 4: Histochemistry of type A trichomes: Nadi reagent. Bar = 25 μm. Figure 5: Secreting cell cytoplasm of type A trichome. Bar = 1 μm. Figures 6, 7: Histochemistry of type C trichome: Fluoral Yellow 088 (6) and FeCl3 (7). Bars = 25 μm. Figures 8, 9: Secreting cell cytoplasms of a young (8) and mature (9) type C trichome. Bars = 1 μm. (Cw) Cell wall; (g) Golgi bodies; (ld) lipidic droplets; (m) mitocondria; (n) nucleus; (p) plastid; (rer) RER; (s) starch; (ser) SER. The essential oil compositions of flowers and leaves differ. The volatile compounds of leaves are characterised by phytol (35.5%), (E)-caryophyllene (19.1%), and hexadecane (7.0%). Oxygenated diterpene hydrocarbons (35.5%), sesquiterpene hydrocarbons (25.0%), and hydrocarbons (22.1%) are the principal fractions. The essential oil of flowers is characterised by six main compounds, (E)-caryophyllene (43.6%), 6,10,14-trimethyl-2pentadecanone (7.5%), 9,12-octadecadienal (5.9%), germacrene D (5.7%), pentadecane (5.7%), and tetradecane (5.6%). Sesquiterpene hydrocarbons (56.9%) and hydrocarbons (19.7%) are the principal fractions. In conclusion, P. majus, considered a typical nonaromatic plant [3,5], bears glandular trichomes which produce small quantities of essential oils, both in leaves and flowers. The plant presents few glandular trichomes on its leaves (type B), but numerous on its inflorescences (types A, B and C). Histochemical observations indicate that the three 2082 Natural Product Communications Vol. 3 (12) 2008 Table 3: Essential oil compositions of leaves and inflorescences of P. majus. RI = Retention Index. Compounds α-Pinene β-Pinene ο-Cymene γ-Terpinene Thymol α-Terpinyl acetate α-Ylangene Tetradecane (E)-Caryophyllene Germacrene D Pentadecane δ-Amorphene Caryophyllene oxide Hexadecane 9,12-Octadecadienal Heptadecane 6,10,14-Trimethyl2-pentadecanone Octadecane Hexadecanol Nonadecane Methyl hexadecanoate Hexadecyl acetate Octadecanol Heneicosane Phytol Docosane Tricosane Tetracosane Pentacosane Total RI Leaves % Flowers % 938 981 1026 1060 1293 1353 1377 1400 1418 1483 1500 1512 1580 1600 1645 1700 0.1 0.1 0.3 0.2 2.7 0.5 1.7 2.7 19.1 2.5 1.4 1.7 7.0 0.9 3.7 5.6 43.6 5.7 5.7 3.9 1.4 4.0 5.9 2.3 1791 1800 1875 1900 1920 2001 2074 2100 2112 2200 2300 2400 2500 2.6 0.2 0.1 1.5 5.1 1.9 35.5 0.5 0.5 1.0 3.5 7.5 1.0 2.1 1.1 3.5 - 93.3% 97.0% types of trichomes produce different kinds of substances (polysaccharides, phenols and essential oil). The organelles observed in the cytoplasm of the secreting cells are consistent with these types of secretion. Essential oil of leaves is produced by type B trichomes, the only type present, considered a typical mucopolysaccharides producer [4]. In this species they are responsible also for the production of the terpenoid fraction, as already observed in Stachys recta [20]. The inflorescences, besides type B hairs, bear other types of trichomes, already described for the Labiatae [18,19]. Type A trichomes have a typical essential oil secretion, while type C trichomes produce a complex secretion, which contains both hydrophilic and lipophilic substances. Essential oil was obtained and analyzed also in flowering plants from Greece [16], but the yield of essential oil is not reported. The composition of Giuliani et al. our samples differs from those of Greece: only α-pinene, γ-terpinene, thymol, (E)-caryophyllene, caryophyllene oxide, and tricosane are present in plants from both sites. Samples A and B from Greece are characterized, respectively, by 1-octen-3-ol (20.7%) and dehydro-aromadendrene (31.8%). The differences could be ascribed not only to the different plant material examined (fresh leaves and flowers in our work, the whole dry plant at flowering time in Greek samples). However, the essential oil composition is certainly affected by the different origin of plant material: the northern part of the distribution area for our samples and typical Mediterranean distribution area for Greek samples. Therefore, the different environmental conditions could be responsible for different chemotypes. It would also be interesting to verify if the plants of southern and warmer regions are richer in essential oil than those of the northern regions. Experimental Plant material: Specimens were collected from two different localities in Tuscany during the blooming period: 03.05.2005 Baratti (Livorno) and 12.05.2007 Giglio Island, Campese (Grosseto). They were determined according to Pignatti [8]. Micromorphological analyses were performed on fresh material (stems, leaves, bracts, calyces and corollas) using scanning electron microscopy (SEM), light microscopy (LM) and transmission electron microscopy (TEM). SEM observations: Small pieces of plant material were fixed in 2.5% glutaraldehyde in 0.1 M phosphate buffer at pH 6.8, dehydrated in ethanol in ascending grades up to absolute and then dried using a critical point dryer apparatus. The samples, coated with gold, were observed with a Philips XL-20 SEM. LM observations: Fresh material was frozen, sectioned and stained using different techniques in order to evidence the different components of the secretion. The stains employed were: Fluoral Yellow088 for total lipids [21], Nile Red for neutral lipids [22], Nadi reagent for terpenes [23], Ruthenium Red [24] and Alcian Blue [25] for acid polysaccharides, and Ferric Trichloride for polyphenols [26]. Observations were made with a Leitz DM-RB Fluo optic microscope. Micromorphology and essential oil analysis of Prasium majus Natural Product Communications Vol. 3 (12) 2008 2083 TEM observations: Small pieces of plant material were fixed in 2.5% glutaraldehyde in 0.1 M phosphate buffer at pH 6.8 and post fixed in 2% OsO4, dehydrated in ethanol in ascending grades up to absolute and embedded in Spurr’s resin. Ultra thin sections were stained with uranile acetate and lead citrate. Samples were examined with a Philips EM300-TEM. capillary columns (30 m x 0.25 mm, 0.25 μm film thickness), working with the following temperature programme: 10 min at 60°C, and subsequently up to 220°C at 5°C/min; injector and detector temperatures, 250°C; carrier gas, helium (1 mL/min); split ratio, 1:20. GC/MS analyses were carried out using an Agilent 5975 GC/MS system operating in the EI mode at 70 eV, using the same columns. The identification of the components was made for both the columns, by comparison of their retention time with respect to n-paraffin (C6-C22) internal standards. The mass spectra and Kovats Indices (KI) were compared with those of commercial (NIST 98 and WILEY) and home-made library mass spectra built up from pure compounds and MS literature data. Isolation and identification of the essential oils: Fresh leaves and inflorescences of the specimen collected at Giglio Island were separately steam distilled for 3 h, in a Clevenger-type apparatus. The essential oil obtained was dried over anhydrous sodium sulfate and stored in sealed vials under refrigeration prior to analysis. Gas chromatography (GC) and gas chromatography-mass spectrometry (GC/MS): The GC analyses were carried out using an Agilent 6890N instrument equipped with HP-WAX and HP-5 Area percentages were obtained electronically from the GC-FID response without the use of either an internal standard or correction factors. References [1] Harley RM, Atkins S, Budantsev AL, Cantino PD, Conn BJ, Grayer R, Harley MM, De Kok R, Krestovskaya T, Morales R, Paton AJ, Ryding O, Upson T. (2004) Labiatae. In: Kadereit JW. (Ed.) The families and genera of vascular plants, vol. 7, Lamiales, Springer, Berlin, 167-282. [2] Cole MD. (1992) The significance of the terpemoids in the Labiatae. In: Harley RM, Reynolds T (eds) Advances in Labiate Science. Royal Botanical Gardens, Kew, 315-324. [3] Werker E. (1993) Function of essential oil-secreting glandular hairs in aromatic plants of the Lamiaceae - A review. Flavour and Fragrance Journal, 8, 249-255. [4] Werker E. (2000) Trichome diversity and development. In: Hallahan DL, Gray JC (Eds.) Advances in Botanical Research. Plant Trichomes. Academic Press, New York-London, 1-35. [5] Hallahan DL. (2000) Monoterpenoid biosynthesis in glandular trichomes of Labiatae plants. In: Hallahan DL, Gray JC (Eds.) Advances in Botanical Research. Plant Trichomes. Academic Press, New York- London, 77-120. [6] Fernandez R. (1972) Prasium L. In: Tutin TG, Heywood VH, Burges NA, Moore DM, Valentine DH, Walters SM, Webb DA. (Eds) Flora Europaea, vol. 3, Cambridge University Press, Cambridge, 137. [7] Greuter W, Burdet HM, Long G. (1986) Prasium L. In: Med-Checklist, vol. 3, Conservatoire et Jardin botaniques de la Ville de Genève, Genève, 312. [8] Pignatti S. (1982). Prasium L. In: Flora d’Italia, vol. 2, Edagricole, Bologna, 448. [9] Martín Mosquero MA, Pastor J, Juan R. (2003) Micromorfología y anatomía de núculas de Prasium majus L. en Andalucía Occidental. Lagascalia, 23, 53-59. [10] Boukamcha H, Jannet HB, Bouazizi Y, Mighri Z. (2003) Isolation and structure determination of a novel furanic ester from the aerial part of Prasium majus. Natural Product Research, 17, 63-66. [11] Kemiss F, Saidane D, Moshtaghie AA. (2005) Effect of aqueous extract of Prasium majus’s L. leaves on water and electrolytes transport in rat intestine. Pakistan Journal of Biological Sciences, 8, 1424-1429. [12] Leonti M, Nebel S, Rivera D, Heinrich M. (2006) Wild gathered food plants in the European Mediterranean: a comparative analysis. Economic Botany, 60, 130-142. [13] Lentini F, Venza F. (2007) Wild food plants of popular use in Sicily. Journal of Ethnobiology and Ethnomedicine, 3:15. Article available from http://www.ethnobiomed.com/content/3/1/15. [14] Mahjoub MA, Jannet HB, Abreu PJM, Mighri Z. (2002) Études biologique et chimique de la plante Prasium majus poussant en Tunisie. Structure d’un heteroside steroidique et d’un steroide. Identification de constituents de la fraction volatile. Journal de la Société Chimique de Tunisie, 4, 1419-1426. [15] Hammami S, Jannet HB, Ciavatta ML, Cimino G, Mighri Z. (2007) A novel iridoid glycoside from the aerial parts of the Tunisian Prasium majus. Natural Product Research, 21, 692-697. 2084 Natural Product Communications Vol. 3 (12) 2008 Giuliani et al. [16] Basta A, Tzakou O, Couladis M, Yannitsaros A. (2007) Essential oil composition of Prasium majus L. from Greece. Flavour and Fragrance Journal, 22, 347-349. [17] Fahn A (2000) Structure and function of secretory cells. In: Hallahan DL, Gray JC (Eds.) Advances in Botanical Research Plant Trichomes. Academic Press, New York-London, 37-75. [18] Corsi G., Bottega S. (1999) Glandular hairs of Salvia officinalis: new data on morphology, localization and histochemistry in relation to function. Annals of Botany, 84, 657-664. [19] Maleci Bini L, Giuliani C. (2006) The glandular trichomes of the Labiatae. A review. Acta Horticulturae, 723, 85-90. [20] Giuliani C, Pellegrino R, Tirillini B, Maleci Bini L. (2008) Micromorphological and chemical characterisation of Stachys recta L. subsp. serpentini (Fiori) Arrigoni in comparison to S. recta subsp. recta (Lamiaceae). Flora, 203, 376-385. [21] Brundrett MC, Kendrick B, Peterson CA. (1991) Efficient lipid staining in plant material with Sudan Red 7B or Fluoral Yellow 088 in polyethylene glycol-glycerol. Biotechnic and Histochemistry, 66, 111-116. [22] Greenspan P, Mayer EP, Fowler SD. (1985) Nile Red: a selective fluorescent stain for intracellular lipids droplets. Journal of Cell Biology, 100, 965-973. [23] David R, Carde JP. (1964) Coloration différentielle des inclusions lipidiques et terpéniques des pseudophylles du Pin maritime au moyen du réactif NADI. Comptes Rendus de l’Academie des Sciences de Paris, 258, 1338-1340. [24] Jensen WA. (1962) Botanical histochemistry: principles and practice, Freeman WH. & Co., San Francisco, 408. [25] Beccari N, Mazzi V. (1966) Manuale di tecnica microscopica, Soc. Editrice Libraria, Como. [26] Gahan PB. (1984) Carbohydrates. In: Plant histochemistry and cytochemistry: an introduction, Academic Press, London, 112-123. NPC Natural Product Communications Olive-oil Phenolics and Health: Potential Biological Properties 2008 Vol. 3 No. 12 2085 - 2088 Francesco Visiolia*, Francesca Ierib, Nadia Mulinaccib, Franco F. Vincierib and Annalisa Romanib a Laboratory of “Micronutrients and Cardiovascular Disease”, UMR7079, UPMC University of Paris 06, Paris, France b Department of Pharmaceutical Science, University of Florence, 50019 Sesto F.no, Florence, Italy francesco.visioli@upmc.fr Received: September 15th, 2008; Accepted: October 29th, 2008 Extra virgin olive oil, the primary source of oil in the Mediterranean diet, differs significantly in composition from dietary lipids that are consumed by other populations. The several minor constituents of virgin olive oil include vitamins such as alphaand gamma-tocopherols (around 200 ppm) and beta-carotene, phytosterols, pigments, terpenic acids, flavonoids, squalene, and a number of phenolic compounds, such as hydroxytyrosol, usually grouped under the rubric “polyphenols”. The antioxidant and enzyme-modulating activities of extra virgin olive oil phenolics, such as their ability to inhibit NF-kB activation in human monocyte/macrophages has been demonstrated in vitro. There is also solid evidence that extra virgin olive oil phenolic compounds are absorbed and their human metabolism has been elucidated. Several activities that might be associated with cardiovascular protection, such as inhibition of platelet aggregation and reduction of plasma rHcy have been demonstrated in vivo. The biologically relevant properties of olive phenolics are described, although further investigations in controlled clinical trials are needed to support the hypothesis that virgin olive oil consumption may contribute to lower cardiovascular mortality. Keywords: extravirgin olive oil, phenolic compounds, Mediterranean diet, hydroxytyrosol. Numerous epidemiological studies have shown that the incidence of coronary heart disease (CHD) and certain cancers, for example breast and colon cancers, is lowest in the Mediterranean basin [1]. It has been suggested that this is largely due to the protective dietary habits of this area [1,2]. The traditional Mediterranean diet, rich in fruit, vegetables, fish, and whole grain, is thought to promote good health and longevity. Olive oil, the primary oil source of this diet, differs significantly in composition from dietary lipids that are consumed by other populations. The formulation of an antioxidant/atherosclerosis hypothesis stimulated experimental and epidemiological studies on the possible role of antioxidants, including olive oil phenolics, in the protection from CHD observed in the Mediterranean area. Included among the minor constituents of virgin olive oil are vitamins such as αand γ-tocopherols (around 200 ppm) and β-carotene, phytosterols, pigments, terpenic acids, flavonoids, squalene, and a number of phenolic compounds, usually grouped under the rubric “polyphenols” [3]. Epidemiological studies: From a nutritional point of view, the choice of a phenol-rich olive oil contributes to the dietary intake of biologically-active compounds in quantities that have been correlated with a reduced risk of developing CHD [4]. Indeed, the use of extra-virgin olive oil as the principle source of dietary oil instead of animal fat, in addition to providing a considerable amount of oleic acid, provides an intake of bioactive compounds with potential healthful effects, as described above. It also appears that the intake and interaction of several "micronutrients" provided by a healthy diet, such as that in use in the Mediterranean area during the mid1940s, is likely to be the link that affords protection from such pathologies [5]. In turn, the answer to the current debate on the efficacy of antioxidant supplements is likely to be found in the adoption of a Mediterranean-style diet, in which the abundance of bioactive, functional compounds provided by fruits, vegetables, wine, and olive oil grants a higher protection toward reactive oxygen species (ROS)induced diseases. 2086 Natural Product Communications Vol. 3 (12) 2008 Visioli et al. In vitro studies: The lower incidence of CHD observed in the Mediterranean area [1] lead to the hypothesis that olive oil phenolics exert a protective effect with respect to chemically-induced oxidation of human LDL, which is one of the initial steps in the onset of atherosclerosis [6]. μM) and are 40-fold weaker than those of the widelyemployed reducing agent ascorbate [13]. Interestingly, two human intervention studies [14,15] confirmed these data in vivo, indicating that extra virgin olive oil might decrease DNA damage, hence lessening cancer risk. Results obtained on human LDL demonstrate that catechol-like compounds present in virgin olive oil inhibit the formation of lipid oxidation products in a dose dependent manner and are effective at a concentration lower than that of pure tyrosol, a phenolic component of the oil, and of probucol, used as reference compounds. This effect is probably due to the synergistic action of hydroxytyrosol, oleuropein aglycones, and some flavonoids, such as quercetin, luteolin, and apigenin present in the virgin olive oil extract in minute amounts. In addition, changes in electrophoretic mobility of apo B are also prevented by the phenols. Oleuropein increases the functional activity of immune-competent cells (macrophages), as demonstrated by a significant increase (58.7 ± 4.6%) in the lipopolysaccharide (LPS)-induced production of nitric oxide, a bactericidal and cytostatic agent [10a]. This increase is consequent to a direct tonic effect of oleuropein on the inducible form of the enzyme nitric oxide synthase (iNOS), as demonstrated by Western blot analysis of cell homogenates and by coincubation of LPS-challenged cells with the iNOS inhibitor L-nitromethylarginine methylester [10b]. Pure hydroxytyrosol (HT) and oleuropein (OE) both potently and dose-dependently inhibit copper sulfateinduced oxidation of LDL at concentrations of 10-6 to 10-4 M [7,8]. The free radical scavenging activities of hydroxytyrosol and oleuropein have been further confirmed [8,9] by the use of metal-independent oxidative systems and stable free radicals, such as DPPH [10a], in a series of experiments that demonstrated both a strong metal-chelation and a free-radical scavenging action. As far as the mechanism of action of olive oil phenolics is concerned, it is well-known that the antioxidant properties of o-diphenols are related to hydrogendonation, which is their ability to improve radical stability by forming an intramolecular hydrogen bond between the free hydrogens of their hydroxyl group and their phenoxyl radicals [10b]. Although specific investigations of olive oil phenols are yet to be carried out, studies performed on the structureactivity relationship of flavonoids indicated that the degree of antioxidant activity is strictly related to the number of hydroxyl substitutions [11]. The mutagenic properties of oxidatively-damaged DNA suggest that antioxidants might have protective activity toward tumor formation. Low concentrations of hydroxytyrosol (50 μM) are able to scavenge peroxynitrite and therefore prevent ONOO-dependent DNA damage and tyrosine nitration [12,13]; also, in a model of copper-induced DNA damage, the prooxidant activities of hydroxytyrosol (due to its copper-reducing properties) become evident at non-physiological concentrations (>500 A correlation between inflammation and cardiovascular diseases has long been established; monocyte/macrophages and NF-κB play a pivotal role. The effects of an extra-virgin olive oil extract, particularly rich in phenolic compounds, were investigated on NF-κB translocation in monocytes and monocyte-derived macrophages (MDM) isolated from healthy volunteers. In a concentrationdependent manner, the extra-virgin olive oil extract inhibited p50 and p65 NF-kB translocation in both unstimulated and phorbol-myristate acetate (PMA)challenged cells, being particularly effective on the p50 subunit. Interestingly, this effect occurred at concentrations found in human plasma after nutritional ingestion of virgin olive oil and was quantitatively similar to that exerted by ciglitazone, a PPAR-γ ligand. However, the extra-virgin olive oil extract did not affect PPAR-γ expression in monocytes and MDM. These data provide further evidence of the beneficial effects of extra-virgin olive oil by indicating its ability to inhibit NF-κB activation in human monocyte/macrophages [16]. In vivo studies: Experimental evidence that phenolic compounds of different origin are absorbed from the diet is accumulating. Animal studies in rats and rabbits demonstrated that LDL isolated from animals fed virgin olive oil exhibit a higher resistance to oxidation when compared with animals given a triglyceride preparation with an equivalent amount of oleic acid, i.e. triolein [17], or «plain» olive oil [18]. We demonstrated that olive oil phenolics are dosedependently absorbed in humans and that they are excreted in the urine, mainly as glucuronide Biological significance of virgin olive oil Natural Product Communications Vol. 3 (12) 2008 2087 conjugates; it is noteworthy that increasing amounts of phenolics administered with olive oil stimulated the rate of conjugation with glucuronide [18]. These data add to the growing experimental evidence that indicates absorption and urinary disposition of flavonoids in humans [19]. The effect of EVOO on platelet aggregation and plasma concentrations of homocysteine (Hcy) redox forms, in relation to the phenolic compounds’ concentration, was also investigated in rats. Three olive oil samples with similar fatty acid, but different phenolic compound concentrations were used: refined olive oil (RF) with traces of phenolic compounds (control oil), native extra virgin olive oil with low phenolic compounds concentration (LC), and extra virgin olive oil with high phenolic compounds concentration (HC) enriching LC with its own phenolic compounds. Oil samples were administered to rats by gavage (1.25 mL/kg body weight) using two experimental designs: acute (24 h food deprivation and killed 1 h after oil administration) and subacute (12 d treatment, a daily dose of oil for 12 d, and killed after 24 h of food deprivation). It is noteworthy that HT exists in the brain as an endogenous catabolite of catecholic neurotransmitters, such as dopamine and norepinephrine [20], but its presence in urine has never, until recently, been described. On the other hand, the formation of homovanillic alcohol (HVAlc), the O-methylated derivative of HT, was reported by Manna et al [21] in human Caco-2 cell incubated with HT. We also reported the urinary excretion of HVAlc, in large excess over its basal excretion (57 ± 3 µg excreted in 24 hours, means ± SD, n= 6). We also described the substrate-induced enhancement of HVA formation, also a product of catecholamines metabolism, in addition to its basal urinary excretion (1660 ± 350 µg excreted in 24 hours, means ± SD, n= 6). Indeed, the results reported suggest that HT increases the basal excretion of HVA, even at the low doses of phenols administered. Future investigations will adopt commercially available virgin olive oils, thus allowing the further elucidation of the in vivo kinetics of olive oil phenolics in habitual consumption quantities. In terms of biological activities, Covas et al. recently reviewed approximately 15 human intervention studies, the vast majority of which indicate that extra virgin olive oil (rich in phenols) is superior to seed oils and olive oil devoid of phenols in modulating selected surrogate markers of cardiovascular disease [22]. One example is an investigation of the effects of olive oil phenols on post prandial events. Bogani et al. evaluated the effects of moderate, real life doses of two olive oils, differing only in their phenolic content, on some in vivo indexes of oxidative stress (plasma antioxidant capacity and urinary hydrogen peroxide levels) in a post prandial setting. Moreover, the authors assessed whether phenolic compounds influence a few arachidonic acid metabolites involved in the atherosclerotic processes, such as leukotriene B4 (LTB4) and thromboxane B2 (TXB2). Six subjects in each group received the three oils [30 mL/day of olive oil (OO), corn oil (CO), or extra virgin olive oil (EVOO), distributed among meals] in a Latin square design. The results demonstrate that EVOO is capable of reducing the post prandial events that associate with inflammation and oxidative stress [23]. Platelet aggregation was induced by ADP (ex vivo tests) and a reduction in platelet reactivity occurred in cells from rats given LC in the subacute study and in cells from rats administered HC in both studies, as indicated by an increase in the agonist half maximal effective concentration. HC inhibited platelet aggregation induced by low ADP doses (reversible aggregation) in cells of rats in both the acute and subacute studies, whereas LC had this effect only in the subacute experiment. Moreover, in rats administered HC in both experiments, the plasma concentration of free reduced Hcy (rHcy) was lower and Hcy bound to protein by disulfide bonds (bHcy) was greater than in RF-treated rats. bHcy was also greater in rats given LC than in RF-treated rats in the subacute experiment. Plasma free-oxidized Hcy was greater in rats given LC and HC than in those administered RF only in the subacute experiment. These results show that phenolic compounds in EVOO inhibit platelet aggregation and reduce the plasma rHcy concentration, effects that may be associated with cardiovascular protection [24]. In conclusion, the biologically relevant properties of olive phenolics described in this article, although still to be further investigated in other controlled clinical trials, provide evidence to support the hypothesis that virgin olive oil consumption may contribute to lower CHD mortality. Acknowledgement – This paper celebrates Professor Vincieri’s birthday. We wish to acknowledge his important contributions to the field. 2088 Natural Product Communications Vol. 3 (12) 2008 Visioli et al. References [1] [2] [3] [4] [5] [6] [7] [8] [9] [10] [11] [12] [13] [14] [15] [16] [17] [18] [19] [20] [21] [22] [23] [24] Keys A. (1995) Mediterranean diet and public health: personal reflections. American Journal of Clinical Nutrition, 61, 1321S-1323S. Hertog MG, Kromhout D, Aravanis C, Blackburn H, Buzina F, Fidanza F, Giampaoli S, Jansen A, Menotti A, Nedeljkovic S, Pekkarinen M, Simic BS, Toshima H, Feskens EJM, Hollman PCH, Katan MB. (1995) Flavonoid intake and long-term risk of coronary heart disease and cancer in the Seven Countries Study [published erratum appears in Archives of Internal Medicine, 155, 1184]. Archives of Internal Medicine, 155, 381-386. Boskou D. (2006) Olive oil: Chemistry and Technology. 2nd Edition. AOCS Press, Urbana, IL, USA. Hertog MLG, Feskens EJM, Katan MB, Kromhout D. (1993) Dietary antioxidant flavonoids and risk of coronary heart disease: the Zutphen Elderly Study. Lancet, 342, 1007-1011. Visioli F, Hagen TM. (2007) Nutritional strategies for healthy cardiovascular aging: focus on micronutrients. Pharmacological Research 55, 199-206. Steinberg D, Parthasarathy S, Carew TE, Khoo JC, Witzum JL. (1989) Beyond cholesterol. Modifications of low-density lipoprotein that increases its atherogenicity. New England Journal of Medicine, 320, 915-924. Visioli F, Bellomo G, Montedoro G, Galli C. (1995) Low density lipoprotein oxidation is inhibited in vitro by olive oil constituents. Atherosclerosis, 117, 25-32. Visioli F, Vincieri FF, Galli C. (1995) 'Waste waters' from olive oil production are rich in natural antioxidants. Experientia, 51, 32-34 Aruoma OI, Deiana M, Jenner A, Halliwell B, Harparkash K, Banni S, Corongiu F, Dessi MA, Aeschbach R. (1998) Effect of hydroxytyrosol found in extra virgin olive oil on oxidative DNA damage and on low-density lipoprotein oxidation. Journal of Agricultural and Food Chemistry, 46, 5181-5187. (a) Visioli F, Galli C. (1998) Olive oil polyphenols and their potential effects on human health. Journal of Agricultural and Food Chemistry, 46, 4292-4296; (b) Visioli F, Galli C. (1998) The effect of minor constituents of olive oil on cardiovascular disease: new findings. Nutrition Reviews, 56, 142-147. Rice-Evans CA, Miller NJ, Paganga G. (1996) Structure-antioxidant activity relationship of flavonoids and phenolic acids. Free Radical Biology and Medicine, 20, 933-956. Aruoma OI, Halliwell B. (1987) Action of hypochlorous acid on the antioxidant protective enzymes superoxide dismutase, catalase and glutathione peroxidase. Biochemical Journal, 248, 973-976. Deiana M, Aruoma OI, Bianchi MLP, Spencer JPE, Kaur H, Halliwell B, Aeschbach R, Banni S, Dessi MA, Corongiu F. (1999) Inhibition of peroxynitrite dependent DNA base modification and tyrosine nitration by the extra virgin olive oil-derived antioxidant hydroxytyrosol. Free Radical Biology and Medicine, 26, 762-769. Weinbrenner T, Fitó M, de la Torre R, Saez GT, Rijken P, Tormos C, Coolen S, Albaladejo MF, Abanades S, Schroder H, Marrugat J, Covas MI. (2004) Olive oils high in phenolic compounds modulate oxidative/antioxidative status in men. Journal of Nutrition, 134, 2314-2321. Salvini S, Sera F, Caruso D, Giovannelli L, Visioli F, Saieva C, Masala G, Ceroti M, Giovacchini V, Pitozzi V, Galli C, Romani A, Mulinacci N, Bortolomeazzi R, Dolara P, Palli D. (2006) Daily consumption of a high-phenol extra-virgin olive oil reduces oxidative DNA damage in postmenopausal women. British Journal of Nutrition, 95, 742-751. Brunelleschi S, Bardelli C, Amoruso A, Gunella G, Ieri F, Romani A, Malorni W, Franconi F. (2007) Minor polar compounds extra-virgin olive oil extract (MPC-OOE) inhibits NF-kappaB translocation in human monocyte/macrophages. Pharmacological Research, 56, 542-549. Scaccini C, Nardini M, D'Aquino M, Gentili V, Di Felice M, Tomassi G. (1992) Effect of dietary oils on lipid peroxidation and on antioxidant parameters of rat plasma and lipoprotein fractions. Journal of Lipid Research, 33, 627-633. Visioli F, Galli C, Bornet F, Mattei A, Galli G, Caruso D. (2000) Olive oil phenols are dose-dependently absorbed in humans. FEBS Letters, 468, 159-160. Wiseman SA, Mathot JN, de Fouw NJ, Tijburg LB. (1996) Dietary non-tocopherol antioxidants present in extra virgin olive oil increase the resistance of low density lipoproteins to oxidation in rabbits. Atherosclerosis, 120, 15-23. Lamensdorf I, Eisenhofer G, White HJ, Hayakawa Y, Kirk K, Kopin IY. (2000) Metabolic stress in PC12 cells induces the formation of the endogenous dopaminergic neurotoxin, 3,4-dihydroxyphenylacetaldehyde. Journal of Neuroscience Research, 60, 552-556. Manna C, Galletti C, Maisto G, Cucciolla V, D’Angelo S, Zappia V. (2000) Transport mechanism and metabolism of olive oil 3,4dihydroxyphenylethanol in CACO-2 cells. FEBS Letters, 470, 341-344. Covas MI. (2007) Olive oil and the cardiovascular system. Pharmacological Research, 55, 175-186. Bogani P, Galli C, Villa M, Visioli F. (2007) Postprandial anti-inflammatory and antioxidant effects of extra virgin olive oil. Atherosclerosis, 190, 181-186. Priora R, Summa D, Frosali S, Margaritis A, Di Giuseppe D, Lapucci C, Ieri F, Pulcinelli FM, Romani A, Franconi F, Di Simplicio P. (2008) Administration of minor polar compound-enriched extra virgin olive oil decreases platelet aggregation and the plasma concentration of reduced homocysteine in rats. Journal of Nutrition, 138, 36-41. NPC Natural Product Communications Traceability of Secondary Metabolites in Eucalyptus and Fagus Wood derived Pulp and Fiber 2008 Vol. 3 No. 12 2089 - 2093 Aline Lamien-Medaa, Karin Zitterl-Eglseera, Heidrun Fuchsb and Chlodwig Franza,* a Veterinärmedizinische Universität Wien, Institut für Angewandte Botanik und Pharmakognosie, Veterinärplatz 1, A-1210 Wien, Austia b LENZING AG, Innovation & Business Development Fiber Science & Entwicklung – TBS, A-4860 Lenzing - Austria Chlodwig.Franz@vu-wien.ac.at Received: July 8th, 2008; Accepted: October 20th, 2008 Industrial pulp and fiber of Eucalyptus and Fagus were investigated for possible identification of secondary metabolites, using chloroform, ethanol and methanol/HCl extracts. The total phenolics test was positive with all the samples and some phenolic compounds like vanillin, vanillic acid and syringic acid were identified by HPLC analysis in the ethanol and methanol-HCl extracts. The extracts had also DPPH radical scavenging activity. Fatty acids like palmitic acid, linoleic acid, oleic acid and stearic acid, cholestane and its derivatives were found in the different extracts by GC/MS analysis. Squalene was also identified and quantified by GC/FID in the dichloromethane extracts. The results showed that the industrial pulp and fibers still contain some secondary plant products comparable to those of the original woods, which confirm the ‘botanical origin’ of the fibers and enables the natural fibers to possess some biological properties, like DPPH antioxidant activity. Keywords: Eucalyptus, Fagus, natural fiber, pulp, phenolic content, antioxidant activity. Natural fibers are those originating directly from nature, usually from plants, animals, and minerals. The common natural fibers are cotton, wool, silk, jute, flax/linen and rock wool. Plant fibers, including vegetable and wood fibers, are generally used in the manufacture of paper and cloth. Several methods are used to convert wood into pulp, including the ground wood, sulfite, and sulfate processes. Other chemical treatments are used to transform the pulp into fiber used for yarn production. Eucalyptus and Fagus woods are two important raw materials used in the textile industry, for example for TENCEL® and Modal® fiber production, respectively. In the pulp and paper industry, extractives from both softwoods and hardwoods cause production and environmental problems. A number of studies have been undertaken on lipophilic wood extractives to determine their composition and to solve pitch problems. Compounds like phenolic acids (ferulic, ellagic, gallic, syringic, vanillic, benzoic and cinnamic acids), aldehydes, tannins, fatty acids, terpenes, sterols, sterol esters, and glycerides have been detected in different types of Eucalyptus and Fagus woods [1-5]. In the present study, phytochemical investigations have been performed on Eucalyptus and Fagus pulp and fiber samples, to see if some secondary plant products are still detectable after the above mentioned industrial processes. HPLC and GC/MS methods have been used for the different extract analyses and the antioxidant activity of the extracts has been tested to evaluate the functionality of the compounds. The total phenolic content of both ethanol and methanol/HCl extracts measured by the FolinCiocalteu assay and expressed in µg caffeic acid equivalents (CAE)/g of dried fiber or pulp, are given in Figure 1. The values varied from 6.5 µg/g with the ethanol extract of Eucalyptus fiber to 16.2 µg/g for the ethanol extract of Eucalyptus pulp. The phenolic content of the ethanol extract of Fagus fiber (9.6 µg/g) was higher than that of Eucalyptus fibers 2090 Natural Product Communications Vol. 3 (12) 2008 Lamien-Meda et al. remaining in the fiber samples, or could also be derived from the partial depolymerization of some residual lignin. Previous studies on Fagus and Eucalyptus woods have shown, however, that phenolic acids like ferulic acid, vanillin, vanillic acid, syringic acid, cinnamic acid derivative, syringaldehyde and resorcilic acid could be identified in Eucalyptus urophylla [3] and E. globulus [1,4,6-8]. Catechin, coniferyl alcohol and sinapyl alcohol have been isolated from the methanol-water extract of Fagus wood [9]. Figure 1: Total phenolic content of ethanol and methanol/HCl extracts of the pulp and fiber samples. GC/MS analysis of the ethanol extract allowed the identification of simple phenolic compounds (vanillic acid, syringic acid), fatty acids (n-tetradecanoic acid, n-pentadecanoic acid, palmitic acid, heptadecanoic acid, linoleic acid, oleic acid, stearic acid), cholestane and its derivatives (cholesterol, cholesta-3,5-dien-7one), and other compounds (xylose, octadecane) (Table 2). These data are in accordance with the literature on the direct wood study of Eucalyptus [1,4,6-8]. The fatty acids identified in Fagus pulp and fiber were also found in Fagus wood [2]. The phenolic compound 2,4-ditert-butylphenol was identified in the ethanol extract of Eucalyptus fiber only (without silylation reaction). This compound was also identified in E. urophylla wood [3] and is well known as an antioxidant compound. 2,4-Ditertbutylphenol has recently been described as an essential oil component of some plants [10-11]. It seems to be useful for the distinction between Eucalyptus and other wood sources, but it is not yet clear if this substance is genuine or an artifact of volatile compounds. (6.5 µg/g). The inverse order was observed with the methanol/HCl extracts: 13.9 and 11.6 µg/g for the Eucalyptus and Fagus fibers respectively. The hydrolyzed extracts (methanol/HCl) had, in general, higher phenolic contents comparatively than the ethanol extracts. Cruz et al. [4] had quantified 1.04 g/100 g (gallic acid equivalent) of total phenolics in Eucalyptus globulus wood, using hydrolyzed extracts and the Folin Ciocalteu assay. This value is thousand times higher than that of Eucalyptus pulp. The high decrease of phenolic content from the wood to the pulp is easy to explain by the industrial process of pulp production. The HPLC/DAD characterization of phenolic compounds in the extracts allowed the identification of vanillin, vanillic acid and syringic acid in both pulp (from wood digestion with magnesium bisulfite acid) and fiber (from the pulp cleaning) extracts (Table 1). In the chromatograms of the ethanol extracts of Eucalyptus pulp and fiber, vanillin, vanillic acid and syringic acid were identified and quantified, but only vanillic and syringic acids were identified in the Fagus fiber. Vanillic acid and syringic acid could be identified in each sample. All the identified phenolic compounds were present in small amounts only (0.3 – 56 µg/10g of pulp or fiber). Squalene was identified and quantified by GC/FID in the dichloromethane extracts of the pulp and fiber of both Eucalyptus and Fagus. The squalene content varied from 2 – 9 µg/10g, with the fiber samples having higher squalene contents than the pulp samples (Figure 2). Squalene was also identified and quantified in E. globulus wood [6,7] at variable concentrations. The The identified and quantified phenolics could be residual compounds from the original wood Table 1: HPLC quantitative results of the pulp and fiber samples. Samples Eucalyptus pulps Fagus pulps Vanillin (µg/10g) Vanillic acid (µg/10g) ETOH MeOH/HCl ETOH MeOH/HCl Syringic acid (µg/10g) ETOH MeOH/HCl 0 – 0.9 0 – 0.6 0.0 – 4.0 0.5 – 1.5 1.1 – 56.0 0.3 – 05.2 0.0 – 2.2 0.4 – 3.8 0.4 – 1.0 0.2 – 1.2 01.9 – 14.2 10.6 – 41.3 Eucalyptus fibers 0 – 0.1 0.0 – 0.2 0 1.3 – 1.7 0.0 – 1.6 43.3 – 52.7 Fagus fibers 0 0.0 0 2.3 0.0 25.0 Phytochemical analysis of wood derived pulp and fiber Table 2: Compounds identified by GC-MS in the ethanol extracts of the Eucalyptus pulp (EP), Fagus pulp (BP), Eucalyptus fiber (EF) and Fagus fiber (BF). RT(mn) 4.675 5.214 7.335 8.254 10.006 11.443 12.329 14.635 16.592 16.799 17.604 17.680 18.063 26.727 31.125 31.243 31.333 31.841 Compounds Butanoic acid Octadecatrienoic acid D-Xylose Vanillic acid Tetradecanoic acid Syringic acid n-Pentadecanoic acid Palmitic acid Heptadecanoic acid Octadecane Linoleic acid Oleic acid Stearic acid Stearic acid derivative Cholesterol Cholestane Cholestane, 2,3-epoxy Cholesta-3,5-dien-7-one EP + + + + + + + + + + + + + + BP + + + + + + + + + + + + + EF + + + + + + + + + + + + + BF + + + + + + + + + + + + + Figure 2: Squalene quantity (GC-FID) in dichloromethane extracts of the pulp and fibers of Eucalyptus and Fagus. Natural Product Communications Vol. 3 (12) 2008 2091 protective activity of squalene against ultraviolet radiation [16] and radiation-induced injury in a mouse model has been demonstrated [17]. The presence of squalene in the final industrial textile should have some protective effect on skin. The presence of active compounds in the pulp and fiber samples was confirmed by testing the antioxidant activity of the ethanol and methanol extracts using DPPH radical scavenging activity. All the extracts had DPPH antioxidant activity and the values ranged from 4.3 – 17.1 µg Trolox equivalents/g sample (Figure 3). A correlation of 0.5 was observed between the total phenolic content and the DPPH antioxidant activity. This positive correlation indicates that the phenolic compounds contributed partly to the antioxidant activity. Indeed, the identified phenolic compounds in the samples (vanillic acid, vanillin, syringic acid) are known to have antioxidant activity. Syringic acid is a more potent DPPH radical-scavenger than BHA and BHT and comparable with ferulic acid, vanillic acid and coumaric acid [18], and presents also antiinflammatory activity [19]. The fatty acids (palmitic, linoleic, oleic, and stearic acids) are known to have antioxidant activity [20] and could contribute to the antioxidant results of the extracts. The present study showed that pulps and fibers originating from wood materials still contain some (functional) secondary plant compounds after the different industrial processing steps. The industrial Eucalyptus and Fagus fibers and pulps contain, for example, small amounts of vanillin, vanillic acid, syringic acid, fatty acids and squalene, which confirm the ‘botanical origin’ of the fibers and enables the natural fibers to possess some biological properties, like DPPH antioxidant activity. Experimental Figure 3: DPPH antioxidant activity of ethanol and methanol/HCl extracts of the pulp and fiber samples. squalene contents (acetone extract followed by a solid phase extraction fractionation) were 1.6 mg/kg [7] and 38.5 mg/kg [6]. Considering the data of [6], the Eucalyptus fiber squalene content was <2 fold lower. Squalene is an isoprenoid molecule with cardioprotective [12], antilipidemic, antioxidant and membrane-stabilizing properties [13-15]. The Fiber samples and sample extractions: Thirteen wood material samples were provided by Lenzing fibers industry, Lenzing/Austria. Lenzing textile general process consists of the transformation of wood to pulp, fiber, yarn and fabric, respectively. The pulp is derived from wood chemical digestion to remove lignin and hemicelluloses, and the fiber derived from the pulp cleaning. The samples analyzed were bleached Eucalyptus pulp (6), produced in craft and sulfite pulping processes, this being the basic material for the production of 2092 Natural Product Communications Vol. 3 (12) 2008 TENCEL®, Fagus pulp (4) from Lenzing Mg-sulfite pulping process, this being the basis of Lenzing Modal®, and further commercial Eucalyptus (2) and Fagus fiber (1) samples. The pulp samples were cut into small pieces using a paper cutter, and normal scissors were used to cut the fiber samples. Three different extracts were prepared from each sample: cold-ultrasonic dichloromethane, reflux with ethanol at 80ºC for 6 h, and reflux with methanol/HCl 2N (1:1) extracts. For each type of extraction, 10 g of sample was extracted with 100-170 mL of solvent. The methanol/HCl extract was neutralized with 9-10 g CaCO3 and partitioned with ethyl acetate (3 x 30 mL), the ethyl acetate part being used. All the extracts were evaporated to dryness under pressure at 40ºC, and dissolved in 1.5 mL methanol for analysis, except those with dichloromethane, which were dissolved in 1.5 mL of dichloromethane for squalene quantification. A blank extract was prepared with each solvent for correction in the different analyses. Spectrophotometric determination of total phenolics: The Folin Ciocalteu reagent was used to determine the total phenolic content [21]. The extracts (40 µL) in 2 mL H2O were mixed with 100 µL of 2N Folin Ciocalteu reagent (Merck, Darmstadt, Germany). This mixture was allowed to stand at room temperature for 3 min and then 200 µL of sodium carbonate (Carl Roth & Co) solution (35 g in 100 mL H2O) was added, and the final volume was completed to 5 mL. After 1 h of incubation in the dark, the absorbance was measured at 725 nm against a water blank using a spectrophotometer (HITACHI 150-20, Ltd. Tokyo, Japan). A calibration curve was plotted using caffeic acid (Sigma-Aldrich Chemie, Steinheim, Germany) (0-40 µg). Determination was performed in duplicate and results were expressed as mg of caffeic acid equivalents (CAE)/ g of dried wool or pulp weight. HPLC determination of phenolic compounds: The wool and pulp extracts were analyzed by HPLC in a Waters instrument fitted with a PDA996 detector, a 626 pump, a 717 plus autosampler and a Symmetry C18 column (5.0 µm, 4.6 x 150 mm) with a column oven temperature at 25°C. Gradient elution was carried out at a flow rate of 1.5 mL/min using 1% acetic acid: acetonitrile 85:15 (solvent A) and methanol (solvent B). The analysis started with 10% B and a linear gradient was used to reach 100% B within 30 min. From 30 to 40 min B was kept constant at 100%. The quantification of vanillin, Lamien-Meda et al. vanillic acid and syringic acid (Sigma-Aldrich Chemie, Steinheim, Germany) was conducted using an external standard (1 – 150 µg/mL) method and detection was at 250 nm. GC-MS and GC-FID analysis: Samples were derivatized as reported by Fukushima and Hatfield [22] with some modification: 1 mL of extract was evaporated to dryness using a rotavapor at 40°C. The dried extract was dissolved in 40 µL of tetrahydrofurane and trimethylsilylated by adding 100 µL of BSA and 10 µL of TMCS. The stoppered tubes with the mixtures were put in an ultrasonicator bath for 5 minutes and kept at 60°C for 30 min. The tubes were cooled to room temperature before GC analysis. The GC/MS (HP 6890 coupled to HP 5972 mass selective detector; Hewlett Packard, Palo Alto, USA) was equipped with a HP-5MS column (length 30 m x 0.25 mm ID, 0.25 µm film thickness; Agilent, Palo Alto, CA, USA), and data were analyzed on a computer equipped with ChemStation software. Helium (average velocity 39 cm/s) was used as carrier gas and the temperature program consisted of an initial temperature of 160°C (held for 5 min), ramp at 4°C/min to 200°C, ramp at 10°C/min to 240°C (held for 5 min), followed by a ramp of 15°C/min to 300°C (held for 10 min). Samples (1 µL) were injected at 250°C and the split ratio was 50:1. Standards (1 mg/mL) of syringic acid, vanillic acid, palmitic acid, linoleic acid, oleic acid, stearic acid, and squalene were used for identification. Squalene (0-171 µg/mL) quantification was carried out using a GC/FID (6890N Network GC system Agilent Technologies, Palo Alto, USA) equipped with a flame ionization detector and a DB-5 narrow bore column (length 10 m x 0.1 mm ID, 0.17µm film thickness; Agilent, Palo Alto, CA, USA). Helium (average velocity 45 cm/s) was used as carrier gas and the oven temperature was increased from 200 to 275°C at 5°C/min, and held for 10 min. The internal standard was 4-androstene-3,17-dione (250 µg/mL). Samples (0.2 µL) were injected at 260°C front inlet temperature and the split ratio was 50:1. DPPH radical scavenging activity: The radical scavenging activity of the sample extracts for the radical 2,2-diphenyl-1-picrylhydrazyl (DPPH, SigmaAldrich Chemie, Steinheim, Germany) was measured as described by Velazquez et al. [23] with some modifications. The extracts (20 µL) were diluted to Phytochemical analysis of wood derived pulp and fiber Natural Product Communications Vol. 3 (12) 2008 2093 100 µL with methanol and mixed with 100 µL of DPPH solution (0.015%). After incubation at room temperature in the dark for 30 min, the absorbance of the reaction mixture was measured at 490 nm using a plate reader (BIO-RAD 450, Japan). Trolox (Fluka, Denmark) (0 – 3.756 µg) was used as standard for the calibration curve. A blank consisting of a high concentration of Trolox (31.3 µg) was used to correct all readings. The results were expressed in mg TE/ g of dried wood or pulp weight. Acknowledgments – This work was financially supported by Lenzing Fiber Industry. References [1] Freire CSR, Silvestre AJ, Neto CP. (2002) Identification of new hydroxyl fatty acids and ferulic acid esters in the wood of Eucalyptus globulus. Holzforschung, 56, 143-149. [2] Zule J, Moze A. (2003) GC analysis of extractive compounds in Fagus wood. Journal of Separation Science, 26, 1292-1294. [3] Huber S. (2004). Charakterisierung der Extraktstoffe von Eucalyptus urophylla. Doctoral thesis, Johannes Kepler Universität Linz. 173p. [4] Cruz JM, Dominguez H, Parajo JC. (2005) Anti-oxidant activity of isolates from acid hydrolysates of Eucalyptus globulus wood. Food Chemistry, 90, 503-511. [5] Conde E, Cadahia E, Garcia-Vallejo MC, Tomas-Barberan F. (2007) Low molecular weight polyphenols in wood and bark of Eucalyptus globulus. Wood and Fiber Science, 27, 379-383. [6] Gutiérrez A, del Rio JC, González-Vila FJ, Martin F. (1999) Chemical composition of lipophilic extractives from Eucalyptus globulus Labill. wood. Holzforschung, 53, 481-486. [7] Rencoret J, Gutiérrez A, del Rio JC. (2007) Lipid and lignin composition of woods from different Eucalyptus species. Holzforschung, 61, 165-174. [8] Esteves B, Graça J, Pereira H. (2008) Extractive composition and summative chemical analysis of thermally treated Eucalyptus wood. Holzforschung, 62, 344-351. [9] Koch G, Puls J, Bauch J. (2003) Topochemical characterization of phenolics extractives in discoloured Fagus wood (Fagus sylvatica L.). Holzforschung, 57, 339-345. [10] Mei W-L, Zeng Y-B, Liu J, Dai H-F. (2007) GC-MS analysis of volatile constituents from five different kinds of Chinese eaglewood. Zhong Yao Cai, 30, 551-555. [11] Rana VS, Blazquez MA. (2007) Chemical constituents of Gynura cusimbua aerial parts. Journal of Essential Oil Research, 19, 21-22. [12] Farvin KHS, Anandan R, Kumar SHS, Shiny KS, Mathew S, Sankar TV, Nair PGV. (2006) Cardioprotective effect of squalene on lipid profile in isoprenaline-induced myocardial infarction in rats. Journal of Medicinal Food, 9, 531-536. [13] Psomiadou E, Tsimidou M. (1999) On the role of squalene in olive oil stability. Journal of Agricultural and Food Chemistry, 47, 4025-4032. [14] Ko T-F, Weng Y-M, Chiou RY-Y. (2002) Squalene content and antioxidant activity of Terminalia catappa leaves and seeds. Journal of Agricultural and Food Chemistry, 50, 5343-5348. [15] Dhandapani N, Ganesan B, Anandan R, Jeyakumar R, Rajaprabhu D, Ezhilan RA. (2007) Synergistic effects of squalene and polyunsaturated fatty acid concentrate on lipid peroxidation and antioxidant status in isoprenaline-induced myocardial infarction in rats. African Journal of Biotechnology, 6, 1021-1027. [16] Kohno Y, Egawa Y, Itoh S. (1995) Kinetic study of quenching reaction of singlet oxygen and scavenging reaction of free radical by squalene in n-butanol. Biochemica et Biophysica Acta, 1256, 52-56. [17] Storm MH, Oh SY, Kimler BF, Norton S. (1993) Radioprotection of mice by dietary squalene. Lipids, 28, 555-559. [18] Von Gadow A, Joubert E, Hansmann CF. (1997) Comparison of the antioxidant activity of aspalathin with that of other plant phenols of Rooibos tea (Aspalathus linearis), α-tocopherol, BHT, and BHA. Journal of Agricultural and Food Chemistry, 45, 632-638. [19] Fernandez MA, Saenz MT, Garcia MD. (1998) Antiinflammatory activity in rats and mice of phenolic acids isolated from Scrophularia frutescens. Journal of Pharmacy and Pharmacology, 50, 1183-1186. [20] Chan P, Juei-Tang C, Chiung-Wen T, Chiang-Shan N, Chuang-Ye H. (1996) The in vitro antioxidant activity of trilinolein and other lipid-related natural substances as measured by enhanced chemiluminescence. Life Sciences, 59, 2067-2073. [21] Singleton VL, Orthofer R, Lamuela-Raventos RM. (1999) Analysis of total phenols and other oxidation substrates and antioxidants by means of Folin–Ciocalteu reagent. Methods in Enzymology, 299, 152-178. [22] Fukushima RS, Hatfield RD. (2001) Extraction and isolation of lignin for utilization as a standard to determine lignin concentration using the acetyl bromide spectrophotometric method. Journal of Agricultural and Food Chemistry, 49, 3133-3139. [23] Velazquez E, Tournier HA, Mordujovich de Buschiazzo P, Saavedra G, Schinella GR. (2003) Antioxidant activity of Paraguayan plant extracts. Fitoterapia, 74, 91-97. NPC Natural Product Communications Potential Anticancer Activity Against Human Epithelial Cancer Cells of Peumus boldus Leaf Extract 2008 Vol. 3 No. 12 2095 - 2098 Juan Garbarinoa, Nicolas Troncosob, Giuseppina Frascac, Venera Cardile c and Alessandra Russod* a Department of Chemistry, University T.F. Santa Maria, Casilla 110-V, Valparaiso, Chile b Lo Vicuña & Cia., www.lo vicuna.cl, Santiago, Chile c Department of Physiological Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy d Department of Biological Chemistry, Medical Chemistry and Molecular Biology, University of Catania, V.le A. Doria 6, 95125 Catania, Italy alrusso@unict.it; ales0303@libero.it Received: July 1st, 2008; Accepted: October 20th, 2008 The potential in vitro antineoplastic effect has been studied of a methanolic extract of leaves of Peumus boldus Molina (Monimiaceae) on two human cancer epithelial cell lines, DU-145 cells (androgen-insensitive prostate cancer cells) and KB cells (oral squamous carcinoma cells). Our findings show that this extract exhibited comparable effects on the cancer cells examined as judged by IC50 values (5.07±0.4 μg/mL and 5.28±0.5 μg/mL in DU-145 and KB cells, respectively). In addition, with respect to genomic DNA damage, determined by Comet assay, the results obtained show a high fragmentation of DNA, not correlated to lactic dehydrogenase (LDH) release, a marker of membrane breakdown, in both cell lines treated with the extract at 5-20 μg/mL concentrations. Taken together, our experimental evidence may justify further investigation of the chemopreventive and chemotherapeutic potential of this natural drug. Keywords: Peumus boldus Molina, DU-145 cells, KB cells, cell growth, DNA fragmentation, LDH release. Boldo consists of the dried leaf of Peumus boldus Molina (Monimiaceae), an evergreen shrub or a small tree growing from central and southern Chile. Boldo, used traditionally in South America mainly against liver diseases, is recognized as a herbal remedy in a number of Pharmacopeias, and is employed in the form of infusions, tinctures and extracts [1,2]. Boldo leaf contains different alkaloids belonging to the large benzylisoquinoline-derived family. Boldine [(S)-2,9-dihydroxy-1,10-dimethoxy-aporphine], the main aporphine alkaloid in boldo leaves and barks, seems to be particularly important as a natural antioxidant [1,2]. Leaves of P. boldus contain also essential oils of complex and variable composition, tannins and flavonoids, such as flavonol glycosides, kaempferol, quercetin and catechin. This last compound is the flavonoid that is most abundant and with the alkaloid boldine is the main contributor of the antioxidant activity of Boldo leaf extracts [3,4]. Cancer is the largest single cause of death in both men and women. Recently, resistance to anticancer drugs has been observed. Therefore, the research and development of more effective and less toxic drugs by the pharmaceutical industry has become necessary. Many substances present in plants, and in particular the flavonoids, are known to be effective and versatile chemopreventive and antitumor agents in a number of experimental models of carcinogenesis [5-8]. In view of these considerations, Boldo leaf could possess anticancer activity and, therefore, could be useful in the prevention or treatment of cancer. The present study was undertaken to investigate the in vitro cytotoxic effect of a methanolic extract from leaves of P. boldus on two human epithelial cancer cell lines, DU-145 cells (androgen-insensitive prostate cancer cells) and KB cells (oral squamous carcinoma cells). To evaluate the effect of the extract from P. boldus leaves on cell growth of human cancer cells, we 2096 Natural Product Communications Vol. 3 (12) 2008 Garbarino et al. Table 1: Cell growth inhibition, assayed using MTT test, of DU-145 and KB cells untreated and treated with methanolic extract from P. boldus leaves at different concentrations for 72 h. DU-145 cells KB cells IC50 a (μg/mL) P. boldus extract 5.07±0.4 5.28±0.5 Doxorubicin 9.37±1.1 1.43±0.7 KB cells Results are expressed as IC50 values (μg/mL) ± SD. The IC50 value, relative to untreated control, represents the concentration that inhibited cell vitality by 50%. Doxorubicin was used as a positive control. Each value represents the mean ± SD of three experiments, performed in quadruplicate. a cultured the cells in either the absence or presence of this natural product. After treatment for 72 h, the MTT assay, a non-radioactive assay widely used to quantify cell viability and proliferation, was performed. The results, summarized in Table 1, show that our extract was active and exhibited comparable degrees of antigrowth effect in both cancer cells examined as judged by IC50 values, 5.07±0.4 μg/mL and 5.28±0.5 μg/mL in DU-145 and KB cells, respectively. Necrosis results in a disruption of the cytoplasmic membrane and the necrotic cells release cytoplasmic lactic dehydrogenase (LDH) and other cytotoxic substances into the medium. We therefore, in a next series of experiments, examined the membrane permeability of treated cells by the existence of LDH in their culture medium. No increase in LDH release was observed in these cancer cells treated with the methanolic extract of P. boldus leaves at 5-10-20 μg/mL concentrations. Conversely, a significant increase (p<0.001) in LDH was observed at 40 μg/mL (Figure 1). Nuclear DNA was analyzed using single-cell gel electrophoresis (SCGE), known as the Comet assay, a sensitive method for the visualization of DNA damage measured at the level of individual cells and a versatile tool that is highly efficacious in human bio-monitoring of natural compounds. The Comet assay also allows us to distinguish apoptotic from normal and necrotic cells based on the DNA fragmentation pattern [9]. The Comet pattern significantly differs between apoptotic and control cultures, as well as between apoptotic and necrotic cultures. Quantification of the Comet data, in our experimental conditions is reported as TDNA and TMOM in Table 2. The results clearly display DNA damage in cells exposed to the P. boldus leaf extract DU-145 cells 80 % LDH release Treatments 100 60 40 * * 20 0 Control 5 10 20 40 μg/ml Figure 1: Lactate dehydrogenase (LDH) release, expressed as percentage of LDH, released into the cell medium with respect to total LDH, in DU145 and KB cells treated with methanolic extract of P. boldus leaves at different concentrations for 72 h. Each value represents the mean ± SD of three experiments, performed in quadruplicate. *Significant vs. control untreated cells (p<0.001). Table 2: Comet assay of genomic DNA of DU-145 and KB cells untreated and treated with methanolic extract of P. boldus leaves at different concentrations for 72 h. Treatments TDNA TMOM DU-145 cells Control 5µg/mL 10 µg/mL 20 µg/mL 40µg/mL 15.7±3.0 99±4.0* 149±6.0* 179±4.3* 30±5.5° 88±3.7 975±15* 1035±12* 1645±43* 287±10* KB cells Control 5µg/mL 10 µg/mL 20 µg/mL 40µg/mL 51±4.0 205±5.0* 298±6.8* 259±3.0* 84±5.9* 175±5.4 1498±16* 2919±14* 2807±13* 337±17* Each value represents the mean ± SD of three experiments, performed in quadruplicate. *Significant vs. control untreated cells (p<0.001). after 72 h, with a drastic increase in both TDNA and TMOM at 5-10-20 μg/mL concentrations. These findings suggest that this extract induces, in our experimental conditions, apoptotic cell death, in accordance with the literature data, which indicate that only comets with high values of TMOM (tail moments) and TD (distance between head and tail of the comet) can be related to apoptosis [10]. Alternatively, in cells exposed to the extract at 40 μg/mL concentration for 72 h, the Comet assay did not show typical comet–like structures that occur during apoptosis. The extract used in the present work could contain boldine, catechin, tannins and other flavonoids and Potential anticancer activity of Peumus boldus Natural Product Communications Vol. 3 (12) 2008 2097 alkaloids, according to reported data [1]. Nevertheless, it not possible from this study to attribute the activity to any of them. However, some suppositions can be advanced on the base of literature data. Flavonoids, kaempferol, quercetin and catechin have been shown to exhibit anticancer activities in preclinical studies [6-8]. The recent studies of Hoet et al. [11] showed that boldine was inactive in a cell growth inhibition assay using HeLa cells (human epithelial cancer cell line from cervical carcinoma). for 72 h under the same conditions. Doxorubicin was used as a positive control. Stock solution of the tested compound was prepared in ethanol and the final concentration of this solvent was kept constant at 0.25%. Control cultures received ethanol alone. Taken together, our experimental data may justify further investigation of the chemopreventive and chemotherapeutic potential of this natural drug. Experimental Chemicals: All reagents were of commercial quality and were used as received. Boldine, catechin, 3(4,5-dimethylthiazol-2-yl)2,5-diphenyl-tetrazolium bromide (MTT) and β-nicotinamide-adenine dinucleotide (NADH) were obtained from Sigma Aldrich Co (St. Louis, USA). All other chemicals were purchased from Sigma Aldrich Co (St. Louis, USA) and GIBCO BRL Life Technologies (Grand Island, NY, USA). Plant material and extraction: The leaves of P. boldo were collected at Quintay (Valparaiso) in January 2006. A voucher specimen (voucher specimen #12-07) was deposited in the Department of Chemistry, Universidad Santa Maria, Valparaiso, Chile. The leaves were exhaustively extracted with methanol and concentrated under vacuum to give a residue [yield 165g (14.3%)]. Cell culture and treatments: DU-145 cells (androgen-insensitive prostate cancer cells) were grown in Dulbecco’s modified Eagle’s medium (DMEM) containing 10% fetal calf serum (FCS), 100 U/mL penicillin, 100 µg/mL streptomycin, 1 mM glutamine, and 1% non essential amino-acids. KB cells (oral squamous carcinoma cells) were maintained in RPMI supplemented with 10% fetal calf serum (FCS), 100 U/mL penicillin, and 100 µg/mL streptomycin. The cells were plated at a constant density to obtain identical experimental conditions for the different tests, thus achieving a high accuracy of the measurements. After 24 h incubation at 37°C under a humidified 5% carbon dioxide atmosphere to allow cell attachment, the cells were treated with different concentrations of methanolic extract of P. boldus leaves, and incubated MTT bioassay: Cellular growth was determined using the MTT assay on 96-well microplates, as previously described [12]. The optical density of each well sample was measured with a microplate spectrophotometer reader (Digital and Analog Systems, Rome, Italy) at 550 nm. Lactic dehydrogenase (LDH) release: Lactic dehydrogenase (LDH) activity was spectrophotometrically measured in the culture medium and in the cellular lysates at 340 nm by analyzing NADH reduction during the pyruvatelactate transformation, as previously reported [12]. The percentage of LDH released was calculated as the percentage of the total amount, considered as the sum of the enzymatic activity present in the cellular lysate and that in the culture medium. A Hitachi U-2000 spectrophotometer (Hitachi, Tokyo, Japan) was used. DNA analysis by Comet assay: The presence of DNA fragmentation was examined by single cell gel electrophoresis (Comet assay), as previously reported [12]. At the end of the electrophoretic run, the “minigels” were neutralized in 0.4 M Tris-HCl, pH 7.5, stained with 100 μL of ethidium bromide (2 μg/mL) for 10 min and scored using a fluorescence microscope (Leica, Wetzlar, Germany) interfaced with a computer. Software (Leica-QWIN) allowed us to analyze and quantify DNA damage by measuring: a) tail length (TL), intensity (TI) and area (TA); b) head length (HL), intensity (HI) and area (HA). These parameters are employed by the software to determine the level of DNA damage as: i) the percentage of the fragmented DNA (TDNA), and ii) tail moment (TMOM) expressed as the product of TD (distance between head and tail) and TDNA. Statistical analysis: Statistical analysis of results was performed by using one-way ANOVA followed by Dunnett’s post-hoc test for multiple comparisons with control. All statistical analyses were performed using the statistical software package SYSTAT, version 9 (Systat Inc., Evanston IL, USA). Each value represents the mean ± SD of three separate experiments performed in quadruplicate. 2098 Natural Product Communications Vol. 3 (12) 2008 Garbarino et al. References [1] Speisky H, Cassels BK. (1994) Boldo and boldine: an emerging case of natural drug development. Pharmacological Research, 29, 1-12. [2] O'Brien P, Carrasco-Pozo C, Speisky H. (2006) Boldine and its antioxidant or health-promoting properties. Chemico-Biological Interactions, 159, 1-17. [3] Schmeda-Hirschmann G, Rodriguez JA, Theoduloz C, Astudillo SL, Feresin GE, Tapia A. (2003) Free-radical scavengers and antioxidants from Peumus boldus Mol. ("Boldo"). Free Radical Research, 37, 447-452. [4] Quezada N, Asencio M, del Valle JM, Aguilera JM, Gómez B. (2004) Antioxidant activity of crude extract, alkaloid fraction, and flavonoid fraction from Boldo (Peumus boldus Molina) leaves. Journal of Food Science, 69, C371-376. [5] Cragg GM, Newman DJ. (2005) Plants as a source of anti-cancer agents. Journal of Ethnopharmacology, 100, 72-79. [6] Leung HW, Lin CJ, Hour MJ, Yang WH, Wang MY, Lee HZ. (2007) Kaempferol induces apoptosis in human lung non-small carcinoma cells accompanied by an induction of antioxidant enzymes. Food Chemical and Toxicology, 45, 2005-2013. [7] Hung H. (2007) Dietary quercetin inhibits proliferation of lung carcinoma cells. Forum Nutrition, 60, 146-157. [8] Bobe G, Weinstein SJ, Albanes D, Hirvonen T, Ashby J, Taylor PR, Virtamo J, Stolzenberg-Solomon RZ. (2008) Flavonoid intake and risk of pancreatic cancer in male smokers (Finland). Cancer Epidemiology Biomarkers & Prevention, 17, 553-562. [9] Bednarek I, Sypniewski D, Klama-Baryła A, Gałka S, Machnik G. (2006) Single-cell gel electrophoresis (comet assay) as a tool for apoptosis determination in tumor cell lines HL-60 and Jurkat cultures treated with anisomycin. Annals Academy of Medicine, 60, 278-284. [10] Godard T, Deslandes E, Lebailly P, Vigreux C, Sichel F, Poul JM, Gauduchon P. (1999) Early detection of staurosporine-induced apoptosis by comet and annexin V assays. Histochemistry and Cell Biology, 112, 155-161. [11] Hoet S, Stévigny C, Block S, Opperdoes F, Colson P, Baldeyrou B, Lansiaux A, Bailly C, Quetin-Leclercq J. (2004) Alkaloids from Cassytha filiformis and related aporphines: antitrypanosomal activity, cytotoxicity, and interaction with DNA and topoisomerases. Planta Medica,70, 407-413. [12] Russo A, Piovano M, Lombardo L, Vanella L, Cardile V, Garbarino J. (2006) Pannarin inhibits cell growth and induces cell death in human prostate carcinoma DU-145 cells. Anti-Cancer Drugs, 17, 1163-1169. NPC Natural Product Communications Antihyperalgesic Effect of Eschscholzia californica in Rat Models of Neuropathic Pain 2008 Vol. 3 No. 12 2099 - 2102 Elisa Vivolia*, Anna Maidecchi b, Anna Rita Biliac, Nicoletta Galeottia, Monica Norcini a and Carla Ghelardinia a Dipartimento di Farmacologia, Università di Firenze, Viale G. Pieraccini 6, 50139 Firenze, Italy b ABOCA SpA Società Agricola, loc. ABOCA 20, 52037 Sansepolcro, (AR) Italy c Dipartimento di Scienze Farmaceutiche, Università di Firenze, Via U. Schiff 6, 50019, Sesto Fiorentino, (FI) Italy elisa.vivoli@unifi.it Received: June 11th, 2008; Accepted: October 23rd, 2008 Eschscholzia californica Cham. (Papaveraceae) is traditionally used by the Indians as a medicinal plant for its anxiolytic, anticonflict, analgesic and sedative properties. The mechanisms of action for the sedative and anxiolytic activities have not been clearly established and so to further investigate the pharmacological profile of E. californica in some painful conditions, a 70% v/v ethanol extract, DERnative=5:1, was tested in rat models of neuropathy induced by chronic constriction injury of the sciatic nerve (CCI), with chemotherapeutic oxaliplatin, and osteoarthritis caused by intrarticular injection of monoiodoacetate. In the CCI model evaluated in the rat paw-pressure test, the examined extract (100 mg kg-1 p.o.) showed an antihyperalgesic effect. Eschscholzia extract, after single injection at a dose of 100-300 mg kg-1 p.o., produced also a statistically significant decrease of pain perception on hyperalgesia induced by oxaliplatin and osteoarthritis, while in the same condition gabapentin did not display any antihyperalgesic effect. Furthermore, in the range of antihyperalgesic doses, the extract was efficacious in the hot-plate (thermal stimulus) and carrageenan tests (inflammatory model) without producing any behavioral impairment, as evaluated by the Irwin test. The analgesic effect exhibited by Eschscholzia extract in the mouse hot-plate test was not antagonized by naloxone, indicating that opioid neurotransmission is not involved in the effect. The above reported results suggest that a 70% (v/v) ethanol dried extract (DERnative=5:1) of E. californica might represent a promising product for the therapy of acute and chronic pain. Keywords: Eschscholzia californica Cham., Papaveraceae, 70% v/v ethanol extract (DERnative=5:1), antihyperalgesic effect, acute and chronic pain. Eschscholzia californica Cham. (California poppy, Papaveraceae) is an annual plant found throughout California and traditionally used by the Indians as a medicinal plant for its anxiolytic, anticonflict, analgesic and sedative properties [1]. The mechanisms of action for the sedative and anxiolytic activities have not been clearly established, although the involvement of several receptors have been observed and recently it has been reported that a 70% (v/v) ethanol extract of California poppy was able to bind to 5-HT(1A) and 5-HT(7) receptors at 100 μg/mL [2]. To further investigate the pharmacological profile of E. californica in some painful conditions, a 70% v/v ethanol extract, DERnative=5:1, was used for the tests in rat models of neuropathy induced by chronic constriction injury of the sciatic nerve (CCI), repeated treatment with the chemotherapeutic agent oxaliplatin, and osteoarthritis caused by intrarticular injection of monoiodioacetate and inflammation caused by carrageenan plantar injection. In the CCI model evaluated in the rat paw-pressure test, the examined extract (100 mg kg-1 p.o.) showed an antihyperalgesic activity (Figure 1). The anti- hyperalgesic effect induced by 300 mg kg-1 did not differ from that obtained with a dose three folds lower (data not shown). California poppy 2100 Natural Product Communications Vol. 3 (12) 2008 Vivoli et al. 70 60 * Weight (g) * * * * 50 40 30 20 CCI OXALIPLATIN MIA CARRAGEENAN 10 0 Sal . Sal . Esch . Gab . 100 100 Sal . Esch . Gab . 100 100 Sal . Esch . Gab . 300 100 Sal . Esch . Gab . 300 100 Figure 1: Effect of Eschscholzia californica extract (ECE) in comparison with gabapentin (Gab.) in rat models of hyperalgesia induced by Chronic Constriction injury (CCI) and by the administration of Oxaliplatin, Monoiodoacetate (MIA) and Carrageenan. Tests were performed 30 min after administration. Doses are expressed as mg kg-1 p.o. *P<0.05 vs saline treated rats. The examined extract, at the dose which was effective in relieving acute and persistent pain, was tested in order to assess its effect on mouse behaviour. At the highest dose employed, the extract did not induce any alteration of either mouse gross behaviour or show any other side effect, as observed in the Irwin test (data not shown). Moreover, mice treated with California poppy extract were evaluated for motor coordination by using the rota-rod test. The endurance time, evaluated before and 15, 30 and 45 minutes after the beginning of the rota-rod test showed the lack of any impairment in the motor coordination of the treated mice (Figure 2). 5 4 Falls in 30 sec extract, after single injection at a dose of 100 mg kg-1 and 300 mg kg-1 p.o., respectively, produced also a statistically significant decrease of pain perception on hyperalgesia induced by oxaliplatin and osteoarthritis, while in the same condition gabapentin did not display any antihyperalgesic effect (Figure 1). Particularly, the extract peaked 30 min after administration (Figure 1). Similarly, in the painful condition caused by intraplantar injection of carrageenan, California poppy extract reduced hyperalgesia at the dose of 300 mg kg-1 p.o., 30 min after administration (Figure 1). Furthermore, in the same range of doses, the extract was efficacious in the mouse hot-plate test in the presence of a thermal stimulus. The analgesic effect exhibited by California poppy extract in the mouse hot-plate test was of an intensity comparable to that exhibited by gabapentin and it was not antagonized by naloxone (1 mg kg-1 i.p.) indicating that opioid neurotransmission is not involved in the effect (Table 1). 3 2 1 0 Pretest 15 30 45 60 min after treatment Figure 2: Effect of Eschscholzia californica extract on mouse rota-rod test: empty square is saline, filled circle is E. californica at a dose of 1000 mg kg-1. Table 1: Effect of Eschscholzia californica extract (ECE) in mouse hot plate test. Licking latency (s) Treatment Pre-test SALINE 14.4±0.5 14.6±0.4 ECE 100 mg kg-1 p.o 14.4±0.7 19.6±0.9* ECE 300 mg kg-1 p.o 14.7±0.7 18.6±0.7* GABAPENTIN 300 mg kg-1 p.o 15.2±0.8 18.6±1.3* NALOXONE 1 mg kg -1 i.p. 14.8±0.5 15.4±0.6 NALOXONE + ECE 100 mg kg-1 p.o. 14.3±0.6 19.7±0.8* *P<0.05 vs saline-treated mice 30 min after treatment Antihyperalgesic effect of Eschscholzia californica The above reported results suggest that the freezedried extract (DERnative=5:1) of E. californica might represent a promising material for the therapy of acute and chronic pain. Natural Product Communications Vol. 3 (12) 2008 2101 Table 2: Mobile phases used for HPLC analysis. Time(min) 0.00 30.00 67 50 B% 33 50 50.00 50 50 1.500 Experimental 65.00 67 33 1.500 Plant material: Eschscholzia californica (California poppy) was cultivated at Aboca’s Organic Farms in 2007 (Aboca, San Sepolcro (AR), Italy). After harvest, the drying process was carried out using special equipment, air-heated ovens, with cells in which forced air is circulated at a temperature and level of humidity that are carefully controlled (the temperature is kept between 32° and 40°C). After this, the desired plant material was separated from any extraneous bodies present (weeds, insects, seeds) based on specific weight, using aero-separators and densimeters, and cut, to reduce the herb to the proper dimensions according to its intended use (for herbal tea, extraction, micronized powder). HPLC-DAD system: The HPLC analyses were performed using a HP 1100 L Palo Alto, CA, USA) equipped with a HP 104iquid Chromatograph (Agilent Technologies, 0 Diode Array Detector (DAD), an automatic injector, an auto sampler, a column oven and managed by a HP 9000 workstation (Agilent Technologies, Palo Alto, CA, USA). Separations were performed on a reversed phase column prodigy 5 μm ODS (3) 100 Å (250.0 × 4.6 mm) column fitted with a 4.0 × 3.0 mm i.d. guard column, both from Phenomenex (Torrance, CA). The eluents were A: SDS 10 mM + DEA 0.1 M in water adjusted to pH 2.5 by H3PO4; B: acetonitrile. The mobile phase is reported in Table 2. The system was operated with an oven temperature of 40oC. Before HPLC analysis, each sample was filtered through a cartridge-type sample filtration unit with a polytetrafluoroethylene (PTFE) membrane [d=13 mm, porosity 0.45 µm (Lida Manufacturing Corp.)] and immediately injected (20 μL). Preparation of plant extract: Eschscholzia californica freeze-dried extract was produced by Aboca Spa (Sansepolcro, AR) and was obtained according to the extraction procedure described below. The dried ground tops were extracted by percolation with a hydro ethanolic solution (70% v/v) and herb/solvent ratio of 1:10. After 8-10 h, the plant extract was filtered to remove the exhausted herb and concentrated under vacuum to remove the ethanol. The extract was freeze-dried under vacuum, without the use of either heat or excipients, at temperatures less than -50°C. The Drug Extract Ratio (DER) was 5:1 and the batch number was n. 7B1150. The content of protopin in this batch was 0.22%, analyzed by a HPLC method. Chemicals: Protopin was purchased from Sigma (Sigma-Aldrich S.r.l., Milan, Italy). All the solvents used for the extraction and HPLC analysis (EtOH, MeOH, diethanolamine and acetonitrile) were HPLC grade from Merck (Darmstadt, Germany). Water was purified by a Milli-Qplus system from Millipore (Milford, MA). The following drugs were used in the pharmacological experiments: Naloxone hydrochloride (Sigma, St. Louis, USA), Oxaliplatin (Sequoia Research Products Ltd, Pangbourne, UK), Sodium iodoacetate (Sigma-Aldrich, Germany). Other chemicals were of the highest quality commercially available. Flow (mL/min) 1.500 1.500 Chromatograms were recorded between 200 and 450 nm. DAD spectra were stored for all peaks exceeding a threshold of 0.1 mAu and detection was performed at 280 nm. Calibration curves: A calibration curve was obtained from an 80% MeOH solution (containing 4% of diluted HCl) of protopin in the range between 0.025 and 0.00625 mg/mL. Pharmacological assays: Drugs were either dissolved in isotonic (NaCl 0.9%) saline solution or dispersed in 1% carboxymethylcellulose sodium salt (CMC, Fluka Chemie GmbH, Steinheim, Germany) immediately before use. Drug concentrations were prepared so that the necessary dose could be administered in a volume of 10 mL kg-1 by i.p. and p.o. injection. Animals: Male Sprague-Dawley albino rats (180-200 g) from Harlan (S.Piero al Natisone, Italy) and male Swiss albino mice (24-26 g) from Morini (San Polo d’Enza, Italy) were used. Four rats and 10 mice were 2102 Natural Product Communications Vol. 3 (12) 2008 housed per cage. For acclimatization, the cages were placed in the experimental room 24 h before the test. The animals were fed a standard laboratory diet and tap water ad libitum and kept at 23±1°C with a 12-h light/dark cycle. All experiments were carried out in accordance with the European Communities Council Directive of 24 November 1986 (86/609/EEC) for experimental animal care. All efforts were made to minimize the number of animals used and their suffering. Chronic constriction injury: A peripheral mono neuropathy was produced in adult rats by placing loosely constrictive ligatures around the common sciatic nerve, according to the method described by Bennet and Xie [3]. Monoiodoacetate injection: Joint damage was induced by a single intra-articular injection of 2 mg of sodium monoiodoacetate into the left knee joint of anaesthetized rats in a total volume of 25 μL. The dose of iodoacetate was chosen based on previous literature [4] and in-house dose response data using 0.5, 1 and 2 mg. Oxaliplatin injection: Hyperalgesia was induced by 15 injections for 5 consecutive days every week for 3 weeks of Oxaliplatin, 2.4 mg kg-1 (15 i.p. injections- cumulative dose 36 mg kg-1 ) [5]. Paw-pressure test: The instrument exerts a force which is applied at a constant rate (32 g per second) with a cone-shaped pusher on the upper surface of the rat hind paw. The force is continuously monitored by a pointer moving along a linear scale. The pain Vivoli et al. threshold is given by the force that induces the first struggling from the rat. Pretested rats which scored below 40 g or over 80 g during the test before drug administration (25%) were rejected. An arbitrary cut off value of 250 g was adopted. Hot plate test: Mice were placed inside a stainless steel container, which was set thermostatically at 52.5±0.1°C in a precision waterbath from KW Mechanical Workshop, Siena, Italy. Reaction times (s) were measured with a stopwatch before and 15, 30, 45 and 60 min after administration of the drug. The endpoint used was the licking of either the forepaws or hind paws. Those mice scoring less than 12 and more than 18 s in the pretest were rejected (30%). An arbitrary cutoff time of 45 s was adopted. Carrageenan test: Rat paw volumes were measured using a plethysmometer (Ugo Basile, Varese, Italy). Rats received the investigated extract 30 min after a 0.1mL injection of 1.0% carrageenan in the right hind paw. Four h after the injection of carrageenan, the pain threshold of the right hind paw was measured and compared with saline/carrageenan-treated controls. Statistical analysis: All experimental results are given as the mean ± S.E.M. An analysis of variance (ANOVA), followed by Fisher’s Protected Least Significant Difference procedure for post-hoc comparison, were used to verify significance between two means. Data were analyzed with the StatView software for the Macintosh. P values of less than 0.05 were considered significant. References [1] Rolland A, Fleurentin J, Lanhers MC, Younos C, Misslin R, Mortier F, Pelt JM. (1991) Behavioural effects of the American traditional plant Eschscholzia californica: sedative and anxiolytic properties. Planta Medica, 57, 212-216. [2] Gafner S, Dietz BM, McPhail KL, Scott IM, Glinski JA, Russell FE, McCollom MM, Budzinski JW, Foster BC, Bergeron C, Rhyu M-R, Bolton JL. (2006) Alkaloids from Eschscholzia californica and their capacity to inhibit binding of [3H]8-hydroxy-2-(diN-propylamino)tetralin to 5-HT1A receptors in vitro. Journal of Natural Products, 69, 432-435. [3] Bennett GJ and Xie YK (1988) A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man. Pain, 33, 87-107. [4] Guingamp C, Gegout-Pottie P, Philippe L, Terlain B, Netter P, Gillet P. (1997) Mono-iodoacetato-induced experimental osteoarthiritis: a dose-response study of loss of mobility, morphology and biochemistry. Arthritis and Rheumatism, 40, 1670-1679. [5] Cavaletti G, Tredici G, Petruccioli MG, Dondè E, Tredici P, Marmiroli P, Minoia C, Ronchi A, Bayssas M, Etienne GG. (2001) Effects of different schedules of oxaliplatin treatment on the peripheral nervous system of the rat. European Journal of Cancer, 37, 2457-2463. NPC Natural Product Communications Problems in Evaluating Herbal Medicinal Products 2008 Vol. 3 No. 12 2103 - 2106 Jozef Corthout Dienst voor Geneesmiddelenonderzoek – Service de Contrôle des Médicaments, Stevinstraat 137, B-1000 Brussels, Belgium corthout.jozef@mail.apb.be Received: June 24th, 2008; Accepted: October 20th, 2008 Compared to chemical drugs, the quality control of herbal medicinal products poses many problems due to the complexity of herbal drugs, herbal drug preparations and the products themselves, and due to the use of different analytical methods (selective and non-selective) giving different results. To illustrate this, three groups of herbal medicinal products were assayed by different analytical methods: products containing St.John’s wort (colorimetry and HPLC), milk thistle (spectrophotometry and HPLC) and ginkgo (HPLC). These studies show that complete and correct labeling is necessary for the evaluation of herbal medicinal products and that for the majority of plants the knowledge of the applied analytical method is essential for a proper verification of conformity. Keywords: Herbal medicinal products, Hyperici herba, Silybi mariani fructus, Ginkgonis folium, analysis. The mission of the Belgian Medicines Control Laboratory is the post-market quality control of medicines delivered in public pharmacies, thus also including herbal medicinal products. Compared with “chemical” drugs, the analysis of herbal medicinal products poses many problems. titrimetry) versus a selective, chromatographic method (HPLC, GC). These problems and differences are illustrated by the evaluation of three groups of herbal medicinal products containing dry extracts of Hyperici herba, Silybi mariani fructus and Ginkgonis folium. First, a plant can be used in different forms: different parts of the plant (for example, leaf, flower, herb); the crude drug itself or different kind of preparations: standardized, quantified, purified or other type of preparation. In case of standardization/quantification this can be performed for either a single constituent or to a group of metabolites. As a consequence of this multiplicity of possibilities, an accurate and detailed label is necessary otherwise it is not possible to verify the product for its conformity. The following information should be stated: Latin name and part of the plant, the ratio of the herbal drug to the herbal drug preparation (DER), the extraction solvent, the physical state, and quantity of the extract. The quantity may also be given as a range corresponding to a defined quantity of constituents with known therapeutic activity [1]. Herbal medicinal products can be on the market as registered medicines or as food supplements, each with their own legal requirements. Finally, the assay of the constituent(s) can give different results depending on the applied method: a non-selective method (spectrophotometry, Hyperici herba or St.John’s wort consists of the whole or cut, dried flowering tops of Hypericum perforatum L., harvested during flowering time, containing not less than 0.08% of total hypericins [2]. The most characteristic constituents are naphthodianthrones, consisting mainly of hypericin and pseudohypericin. Other naphthodianthrones present, and included in the term ‘total hypericin’, are the biosynthetic precursors protohypericin and protopseudohypericin (which are transformed into hypericin and pseudohypericin, respectively on exposure to light), and a small amount of cyclopseudohypericin. Other constituents include phloroglucinols and flavonoids. St. John’s wort is indicated for mild to moderate depressive episodes in daily dosages of 450 to 1050 mg of hydroalcoholic dry extract (with defined DER) [3]. Eight St. John’s wort preparations (tablets, coated tablets or capsules) were analyzed by assaying total hypericins by spectrophotometry [2] and HPLC [4]. None of the products had a comprehensive label, the 2104 Natural Product Communications Vol. 3 (12) 2008 Corthout Table 1: Products containing St. John’s wort. Prod 1 2 3 4 5 6 7 8 Label claim mg TH/unit mg extract/unit 0.3 NM 0.3 NM 0.3 NM 0.4 NM 0.4 NM 0.5 100-120 0.9 250-300 0.85 425 Spectrophotometry mg TH/unit % of claim 0.21 70.0% 0.20 60.6% 0.27 90.0% 0.34 85.0% 0.08 26.7% NA interference 0.79 87.8% 1.5 176.5% HPLC mg TH/unit % of claim 0.11 36.7% 0.11 33.3% 0.05 16.7% 0.13 32.5% 0.04 13.3% 0.13 26.0% 0.38 42.2 0.72 84.7% H, Hypericin, NM, Not Mentioned TH, Total Hypericins, NA, Not Applicable most common missing information being part of the plant, type of extract, solvent, DER and even the quantity of the extract (5 products). Five products were quantified to a defined content of hypericin, two to total hypericin and one to hypericinum (?). One product gave a minimum content of hypericin. The assay results are summarized in Table 1: the total hypericin content varied from 26.7% to 176.5% of their label claim for the spectrophotometric assay and from 13.3% to 84.7% for the HPLC method. The ratio of the spectrophotometric assay to the HPLC assay ranged from 2.1 to 5.4, which is in accordance with the results of the group of Prof. Vincieri and of Dr Gaedke, who obtained for commercial dry extracts ratios of 1.2 to 10 and 1.1 to 1.6, respectively [5,6]. None of the products complied with its label claim neither for the spectrophotometric result nor for the HPLC result, taking 90.0%-110.0% of the stated amount as requirement. Silybi mariani fructus (Cardui mariae fructus) or milk thistle fruit is the mature fruit, devoid of the pappus, of Silybum marianum (L.) Gaertner, containing a minimum of 1.5% of silymarin, expressed as silibinin. Milk thistle refined and standardized dry extract contains 30 to 65% of silymarin (determination by HPLC). The content of silymarin corresponds to 20-45% silicristin and silidianin, 4065% silibinin A and B, and 10-20% isosilibinin A and B, with reference to total silymarin [7]. The active constituents are flavanolignans, collectively named silymarin, and consisting mainly of silibinin and isosilibinin. The milk thistle dry extract, which is the most frequently used in the pharmacological/ Table 2: Products containing milk thistle. Prod 1 2 Label claim mg S/caps mg extract/caps 140 180 200 250 Colorimetry mg S/caps % of claim 141.6 101.1% 165.2 82.6% 3 451 NP 4 75 250 138.7 184.9% HPLC mg S/caps % of claim 110.9 79.2% 115.5 57.8% 45.5 101.1% 122.3 163.1% 1 contains silibinin as silibinin phosphatidylcholine S, silymarin; NP, Not Performed toxicological and clinical studies, contains 74.180.9% of silymarin (determination by UV spectroscopy; DER 36-44:1) [8]. Milk thistle products are used in cases of toxic liver damage; chronic inflammatory liver conditions and hepatic cirrhosis in daily dosages equivalent to 154-324 mg of silymarin (HPLC method) or 200-420 mg of silymarin (colorimetric method). The silymarin content obtained by HPLC amounts to about 77% of the value obtained by colorimetry [8]. Four milk thistle preparations, all formulated as capsules, were evaluated by determination of the silymarin content by spectrophotometry [9] and HPLC [7]. Three products contained a dry extract standardised to different amounts of silymarin; one product contained pure silibinin. The results are summarized in Table 2: for the silibinin product, 101.1% of the label claim was found by HPLC, for the other products the silymarin content varied from 82.6% to 184.9% of their label claim for the colorimetric assay and from 57.8% to 163.1% for the HPLC method. The ratio of the HPLC assay result to the colorimetric assay result ranged from 0.7 to 0.9, which is in agreement with the results of Ihrig [9], where the same ratio range was obtained. Products 1 and 3 complied with their label claim taking the content from the colorimetric analysis and the HPLC analysis, respectively. Despite the fact that products 2 and 4 are food supplements, they contain silymarin in either the same or higher amount as the medicinal product 1. Ginkgonis folium or ginkgo leaf is the whole or fragmented, dried leaf of Ginkgo biloba L., containing not less than 0.5% of flavonoids, calculated as flavone glycosides. Ginkgo refined and quantified dry extract contains 22.0-27.0% flavonoids (determined by HPLC-UV), 2.6-3.2% bilobalide and 2.8-3.4% ginkgolides A, B and C (determined by HPLC-RI) [10]. The characteristic constituents of Problems in evaluating herbal medicinal products Natural Product Communications Vol. 3 (12) 2008 2105 ginkgo leaf are terpenes and flavonoids. The principal terpenes are diterpene trilactones called ginkgolides and the sesquiterpene trilactone bilobalide. The main flavonoids are mono-, di- and triglycosides of the flavonols quercetin, kaempferol and isorhamnetin. Diglycosides esterified with p-coumaric acid are also present. Other flavonoids include flavones, biflavones, flavan-3-ols and proanthocyanidins. Ginkgo is indicated for mild to moderate dementia syndromes including primary degenerative dementia, vascular dementia and mixed forms; cerebral insufficiency; neurosensory disturbances such as dizziness, vertigo and tinnitus; enhancement of cognitive performance; and peripheral arterial occlusive disease (intermittent claudication). All these indications are supported by clinical trials performed with preparations based on standardized extracts, containing 22-27% flavonol glycosides and 5-7% terpene lactones. The daily dose for these preparations is 120-240 mg of standardized ginkgo dry extract, corresponding to 29-58 mg flavonoids and 7.2-14.4 mg terpene lactones [11]. between 8 mg and 44.5 mg total flavonoids per unit. Generally the ratio of the 3 aglycones quercetin, kaempferol and isorhamnetin in ginkgo is 5:5:1, corresponding to a relative rutin content of 10 to 15%. Because it is known that rutin is sometimes added to ginkgo extracts, a direct assay of rutin was also performed [12]. Absolute and relative contents (with reference to total flavonoids) ranged from 0.4 to 8.9 mg/unit and from 5 to 51%, respectively. Products 4-7, 11, 13 and 14 had a relative rutin content ranging from 24% to 51%, indicating a possible falsification with rutin. A total of 14 products, formulated as capsules and tablets, were included in the study (see Table 3). They all claim to contain ginkgo dry extract, ranging from 30 to 200 mg per unit. For ginkgo, the problem of different existing assay methods is not a point because in most cases the flavonoids are determined with HPLC-UV and the terpene lactones with HPLC-RI [12]. The content of total flavonoids was determined after hydrolysis to the flavonol aglycones quercetin, kaempferol and isorhamnetin. Eleven of the 14 products had a claim for flavonoid content ranging from 9.6 mg up to 36 mg per unit. For these products, between 95% and 123% of the theoretical amount was found. For the 3 other products without indication of flavonoid amount, the content varied Concerning the analysis of terpene lactones, for products 1-8 having a label claim, the assayed amount varied from 80% (product 4) up to 194% (product 8) of the theoretical amount. Products 9-14 (without a claim) had a content of terpene lactones between 1.42 and 4.83 mg per unit. Only the medicinal products 1-3 complied with all the requirements: the content of total flavonoids and terpene lactones were in accordance with the stated amounts; the relative rutin content was between 10 and 15% of the total flavonoid content and their label was complete and correct. Again, for most of the food supplements the content and the corresponding daily dose of flavonoids and terpene lactones was higher when compared with the medicinal products: product 9 had no label claim, but its daily dose for flavonoids and terpene lactones corresponded respectively to about 460% and 200% of a normal daily dose. These studies show that a complete and correct label is necessary for the evaluation of herbal medicinal products. In addition, for the majority of plants, the knowledge of the applied analytical method is essential to enable a proper verification of the conformity. Table 3: Products containing ginkgo. Prod 1 2 3 4 5 6 7 8 9 10 11 12 13 14 extract/unit 40 mg 60 mg 40 mg 60 mg 50 mg 80 mg 100 mg 60 mg 200 mg 150 mg 60 mg 63 mg 200 mg 30 mg Label claim flavonoids/ terpene unit lactones/unit 9.6 mg 2.4 mg 14.4 mg 3.6 mg 9.6 mg 2.4 mg 14.4 mg 3.6 mg 12.0 mg 3.0 mg 19.2 mg 4.8 mg 24.0 mg 6.0 mg 14.4 mg 3.6 mg NM NM 36.0 mg NM 14.4 mg NM 15.0 mg NM NM NM NM NM NM, Not Mentioned, NA, Not Applicable Total Flavonoid Assay A: flavonoids/unit B: % of claim 9.14 mg 95.2% 14.53 mg 100.9% 9.23 mg 96.1% 16.97 mg 117.9% 13.45 mg 112.1% 23.63 mg 123.1% 23.02 mg 95.9% 14.70 mg 102.1% 44.50 mg NA 35.25 mg 97.9% 16.34 mg 113.4% 15.28 mg 101.9% 13.21 mg NA 7.98 mg NA Rutin Assay A: rutin/unit B: % of total flavonoids 1.05 mg 11% 1.57 mg 11% 1.17 mg 13% 4.19 mg 25% 6.82 mg 51% 8.89 mg 38% 5.44 mg 24% 1.54 mg 10% 4.99 mg 11% 3.41 mg 10% 5.50 mg 34% 1.70 mg 11% 3.60 mg 27% 2.89 mg 36% Terpene lactone Assay A: terpene lactones/unit B: % of claim 2.39 mg 99.6% 3.71 mg 103.1% 2.31 mg 96.3% 2.88 mg 80.0% 4.27 mg 142.3% 6.10 mg 127.1% 5.25 mg 87.5% 6.98 mg 193.9% 4.83 mg NA 4.63 mg NA 3.93 mg NA 4.56 mg NA 3.64 mg NA 1.42 mg NA 2106 Natural Product Communications Vol. 3 (12) 2008 The analysis of ginkgo, showing a possible falsification of ginkgo extract by adding rutin, demonstrates the benefit of using a chromatographic assay method. An additional remark is that most of the examined food supplements do not comply with either their label claim or the defined requirements and that their content is sometimes higher than the minimum therapeutic dose ascribed to the medicinal products. Experimental Materials and apparatus: Herbal drug preparations were purchased from a local wholesaler. The following standards were used: hypericin CRS from the European Pharmacopoeia, silibinin and quercetin trihydrate from Sigma, rutin and standardized ginkgo dry extract (1.27% ginkgolide A, 0.77% ginkgolide B, 1.32% ginkgolide C and 2.65% bilobalide) from Wilmar Schwabe. The UV-VIS analyses were performed on an Agilent 8453 UV-VIS spectrophotometer. The HPLC analyses were carried out on either a Waters Alliance 2695 equipped with a 2996 PDA detector or a Waters 410 Differential Refractometer. Chromatographic conditions: Hypericin assay: The column was a Symmetry C18 250 x 4.6 mm, 5 µm (Waters), used at ambient temperature. The samples were chromatographed with a mixture of 150 mL buffer pH 2.1 (13.98 g NaH2PO4.1H2O in 1000 mL water adjusted to pH 2.1 with H3PO4 85%), 140 mL ethyl acetate and 760 mL methanol as mobile phase, with detection at 590 nm, a flow rate of 1.0 mL/min and 20 µL as injection volume. Corthout Silymarin assay was carried out on a Waters Symmetry C18 150 x 4.6 mm, 5 µm column at ambient temperature using a gradient at 1.0 mL/min. with 2 solvents (A = H3PO4, methanol, water (0.5:35:65); B = H3PO4, methanol, water (0.5: 50:50); 100%A/0%B to 0%A/100%B in 28 min and continuing this for 8 min). The injection volume was 10 µL and UV detection was at 280 nm. Total flavonoid assay utilized a Waters Symmetry C18 150 x 4.6 mm, 5 µm column held at 25°C in combination with a gradient at 1.0 mL/min. using 2 solvents (A = 0.3 g/L of H3PO4 in water adjusted to pH 2.0; B = methanol; 60%A/40%B to 45%A/55%B in 20 min.). The injection volume was 10 µL and UV detection was at 370 nm. Rutin assay was carried out on a Varian Polaris C18 250 x 4.6 mm 5 µm column at ambient temperature using a mixture of 3 g/L of H3PO4 in water and acetonitrile (82:18) as mobile phase, at a flow rate of 1 mL/min, injection volume of 10 µL and UV detection at 357 nm. Terpene lactone assay: the mobile phase, a mixture of water– methanol–tetrahydrofuran (75–20–10) was pumped at 1.0 mL/min through a Waters Symmetry C8 250 x 4.6 mm, 5 µm column at 35°C, followed by a refractometer, also at 35°C. The injection volume was 100 µL. Quantitative determination: The samples were prepared as described in the cited references. Each determination was performed in triplicate except the terpene lactone HPLC assay, which was undertaken in duplicate. The contents are presented as a mean of data obtained from the spectrophotometric and HPLC analyses. References [1] [2] [3] [4] [5] [6] [7] [8] [9] [10] [11] [12] Guideline on quality of Herbal Medicinal Products/Traditional Herbal Medicinal Products (CPMP/QWP/2819/00 Rev1). St.John’s wort – Hyperici herba. (2008) In: European Pharmacopoeia 6.0. St.John’s wort – Hyperici herba. (2003) In: ESCOP Monographs, The scientific foundation for herbal medicinal products, 78-210. Krämer W, Wiartalla R. (1992) Bestimmung von Naphtodianhronen (Gesamthypericin) in Johanniskraut (Hypericum perforatum L.). Pharmazeutische Zeitung Wissenschaft, 5, 202-207. Bergonzi MC, Bilia AR, Gallori S, Guerrini D, Vincieri FF. (2001) Variability in the content of the constituents of Hypericum perforatum L. and some commercial extracts. Drug Development and Industrial Pharmacy, 27, 491-497. Gaedke F. (1997) Johanniskraut und dessen Zubereitungen. Deutsche Apotheker Zeitung, 137, 3753-3757. Milk thistle fruit – Silybi mariani fructus. Milk thistle dry extract, refined and standardised – Silybi mariani extractum siccum raffinatum et normatum. In: European Pharmacopoeia 6.0. (2008) Blaschek W, Ebel S, Hackenthal E, Holzgrabe U, Keller K, Reichling J, Schulz V. (Eds) (2007) Silybum. In: Hagers Enzyklopädie der Arzneistoffe und Drogen 6th ed., Band 14, WVG Stuttgart, 582-605. Ihrig M, Meiss M, Möller H. (1999) Gehaltbestimmung von Silymarin in Fertigarzneimitteln. Pharmazeutische Zeitung, 34, 2661-2671. Ginkgo Leaf – Ginkgonis folium. Ginkgo dry extract, refined and quantified – Ginkgonis extractum siccum raffinatum et quantificatum. In: European Pharmacopoeia 6.0. Ginkgo Leaf – Ginkgo folium. (2003) In: ESCOP Monographs, The scientific foundation for herbal medicinal products, 178-210. Saevels J, Corthout J. (2005) Ginkgo biloba medicines and food supplements. Journal de Pharmacie de Belgique, 4, 129-134. NPC Natural Product Communications Impurities in Herbal Substances, Herbal Preparations and Herbal Medicinal Products, IV. Heavy (toxic) Metals* 2008 Vol. 3 No. 12 2107 - 2122 SFSTP Commission, Didier Guédona, Michèle Brumb, Jean-Marc Seigneuretc, Danièle Bizetd, Serge Bizote, Edmond Bournyf, Pierre-Albert Compagnong, Hélène Kergosienh, Luis Georges Quintelasi, Jerôme Respaudj, Olivier Saperask, Khalil Taoubil and Pascale Urizzim a Laboratoires Arkopharma, ZI Carros Le Broc, BP 28, F-06511 Carros Cedex, France 48, avenue de la République, F-92500 Rueil-Malmaison, France c Alban Muller, 8, rue Charles-Pathé, F-94300 Vincennes, France d Laboratoires Boiron, 20, rue de la Libération, F-69110 Sainte-Foy-les-Lyon, France e Indena, 38, avenue Gustave-Eiffel, F-37095 Tours, France f LPPAM, avenue de la Gare, BP 47, F-26170 Buis-les-Baronnies France g AFSSAPS, 143-147, boulevard Anatole-France, F-93285 Saint-Denis, France h Euromed France, 95, route du Morgon, F-69400 Gleizé, France i SQUALI, 3, rue de la Pagère, F-69500 Bron, France j Avogadro, Parc de Genibrat, F-31470 Fontenilles, France k LACAPA, 3, boulevard de Clairfont, F-66350 Toulouges, France l Laboratoires Boiron, 20, rue de la Libération, F-69110 Sainte-Foy-les-Lyon, France m Institut de Recherche Pierre Fabre, 3, rue Ariane, BP 72101, F-31521 Ramonville-Saint-Agne Cedex, France b didier.guedon@arkopharma.com Received: July 29th, 2008; Accepted: October 23rd, 2008 The main source of available forms of heavy metals (toxic metals) for the plant kingdom is anthropogenic, resulting from diverse activities such as metallurgic processing of ore, cement plants, uncontrolled discharge of sewage sludge, burning of fossil fuels and waste incineration plants, and leaded petrol. Agricultural chemicals (e.g. phosphate fertilizers containing cadmium) may also contribute to the contamination of cultivated plants. The main threats to human health from toxic metals are associated with exposure to lead, cadmium, mercury (organic forms, especially methylmercury) and arsenic (mineral form only), which have no known vital or beneficial effect on living organisms. As their toxicity often takes years to manifest and may go unsuspected, their toxicological risk is defined on the basis of the so-called Provisional Tolerable Weekly Intake (PTWI) values. Beside anthropogenic causes, the main factors that may lead to high levels of toxic metals in medicinal plants are their availability in the soil with soil pH as the most important parameter for uptake by the plant. Indeed, genetic features of certain plant species show a tendency to accumulate certain trace elements, especially cadmium (“cadmium collector”). A very recent revision draft of the monograph “Herbal drugs” (Ph. Eur., 1433) includes acceptance criteria for lead, cadmium and mercury. This proposal is discussed in detail, based on literature data dealing with terrestrial plants and seaweed. Additionally, the need for inclusion of tests for inorganic impurities in quality control specification is examined, based on a risk assessment. As the daily intake of food supplements is very similar to the one of herbal remedies, it would be advisable to take into account the same acceptance criteria. The specific situation has also been considered of exotic herbal remedies, particularly those of Asian origin, which have been repeatedly reported to contain toxic levels of heavy metals and/or arsenic resulting in heavy metal poisoning. Keywords: Herbal drugs, herbal drug preparations, herbal medicinal products, food supplements, heavy metals, toxic metals, quality control, regulations. The term “heavy metals” has usually been used to ________________________________________________________________________________________________________________________________________________________________________________________ * This publication pertains to an article with several parts. Parts I (“Microbial contamination”), II (“Mycotoxins”) and III (“Pesticide residues”) have already been published [1-3]. describe elements that should be restricted in ingested materials because of their toxic effects. Based on physical properties, this term includes not only lead 2108 Natural Product Communications Vol. 3 (12) 2008 and cadmium but also other elements between chromium and bismuth on the periodic table of the elements, having specific gravities greater than 5 g/cm3. Some of them are essential nutrients in trace amounts (e.g. copper, iron, zinc, cobalt, manganese, molybdenum) and others have relatively low toxicity (e.g. nickel, chromium). Mercury, a noble metal, is also an element that is controlled in certain foods because of its toxic nature. Arsenic, a metalloid (usually classified as a heavy metal), may be of concern. So, the term “toxic metals” is presently most often used in the literature, unlike the European Pharmacopoeia (Ph. Eur.), which prefers so far the wording “heavy metals”. Both of them will be used in the present publication, along with trace element. Heavy metals, as natural components of the earth surface (minerals, ore), do not present any risk for human health as they are not available for living organisms. They are released into the environment due to many factors including natural (e.g. erosion or volcanic activity) and mainly anthropogenic causes, for example, metallurgical processing of ore (mining industry, smelting works), cement plants, uncontrolled discharge of sewage sludge, contaminated emissions from refineries, burning of fossil fuels or waste incineration plants, and leaded petrol. Heavy metal uptake by plants from the soil is quite variable. Root systems and soil properties, such as pH, may determine their availability to the plant (higher uptake in acid soils due to higher solubility of heavy metals) [4,5]. The level of lead, cadmium and other metallic trace elements in a raw material can vary considerably with plant part, habitat and soil concentration [6-8]. Industrial activities have been identified as responsible for abnormally high accumulations of lead and cadmium in medicinal plants [6,9,10]. Meanwhile, heavy metals content in a soil is not necessarily a sufficient indicator to evaluate their possible accumulation in plants [11] as stronger genetic features may also influence heavy metals content [6,12-14]. Some plants have been recommended for cleaning toxic metals from polluted soils due to their strong absorption and concentration from the soil [6]. There are two major reasons why it has become necessary to consider levels of toxic metals in herbal drugs and/or their preparations and finished products: • contamination of the environment with toxic metals has increased dramatically during the 20th century, especially cadmium emissions while, Guédon et al. over the last decades, lead emissions in developed countries have decreased markedly due to the introduction of unleaded petrol [4,15]. The sources of this environmental pollution are quite varied, ranging from industrial activities (see above) to the use of purification mud and agricultural treatments, such as cadmiumcontaining phosphate fertilizers, mercury fungicides and arsenical insecticides, much used some years ago and even used today in some countries [8,16]. It is also generally agreed that cadmium in fertilizers is by far the most important source of cadmium input to soil and to the food chain. Cadmium accumulation in agricultural soils due to fertilizer application was the subject of an opinion delivered by SCTEE [17]. As an example, cadmium, lead and mercury were significantly higher in the roots from valerian grown on sewage sludge-amended soil, than on unamended soil [18]; • exotic herbal remedies, particularly those of Asian origin, have been repeatedly reported to contain toxic levels of heavy metals and/or arsenic [19]. 1. PRESENCE OF HEAVY METALS DUE TO ENVIRONMENTAL POLLUTION 1.1. Cadmium and lead: Most of the studies on the presence of toxic metals in medicinal plants have focused on lead and cadmium [5-8,12,13,15,20-30]. The contamination of herbal drugs with lead and cadmium has been shown to be subject to broad fluctuations depending on the plant species. A German working group on contaminants, running a data base on heavy metals, published a compilation of results. A total of over 12,000 samples corresponding to 204 herbal drugs were tested for lead and cadmium content, comparing minimum and maximum values, along with the 90th percentile (i.e. level below which 90% of the findings occur) [14]. The 90th percentile of many herbal drugs was over 0.5 mg/kg of cadmium (and even over 1.0 mg/kg), while far fewer raw materials showed a 90th percentile exceeding 5 mg/kg of lead (see Table 1). The Official Medicines Control Laboratories (OMCL) network of the European Directorate for the Quality of Medicines & Healthcare (EDQM) conducted a market surveillance study regarding cadmium content of different herbal drugs from the European market [31]. The main results are presented, with the kind permission of EDQM. Toxic metals in herbal medicinal products Natural Product Communications Vol. 3 (12) 2008 2109 Table 1: Cadmium and lead: 90th percentile in herbal drugs [14]. Heavy metals Cadmium: Herbal drugs Speedwell (0.52 mg/kg), Celery, fruit (0.53 mg/kg), Sundew (0.53 mg/kg), Ivy (0.53 mg/kg), Lady’s mantle (0.54 mg/kg), Dill, fruit (0.54 mg/kg), Linseed (0.54 mg/kg), Dill, herb (0.58 mg/kg), Woodruff (0.59 mg/kg), Kava-kava (0.63 mg/kg), Lemon balm (0.64 mg/kg), Dandelion, herb and root (0.64 mg/kg), Birch (0.67 mg/kg), Immortelle (0.69 mg/kg), Dandelion, herb (0.69 mg/kg), Lungwort (0.79 mg/kg), Buckwheat (0.86 mg/kg), True golden rod (0.86 mg/kg), Spinach (0.93 mg/kg), Wild pansy (1.00 mg/kg), Bladder wrack (1.05 mg/kg), Fumitory (1.05 mg/kg), Mallow, leaf (1.17 mg/kg), St John’s wort, herb (1.30 mg/kg), St John’s wort, flower (1.43 mg/kg), Willow (1.80 mg/kg), Tormentil (2.13 mg/kg). 90th percentile ≥ 0,5 mg/kg Clove (5.3 mg/kg), Sundew (6.6 mg/kg), Buckthorn (7 mg/kg), Ginkgo (10.5 mg/kg), Mallow, leaf (12 mg/kg), Island moss (14.3 mg/kg). Lead: 90th percentile ≥ 5,0 mg/kg Table 2: Cadmium in herbal drugs: herbal drugs with a cadmium content higher than 0.3 mg/kg [31]. Yarrow, aerial parts 8 Number of batches Cd > 0.3 mg/kg 7 Birch, leaf 12 11 8 --- Oak, bark German chamomile, flower h d Mallow, flower 7 3 --- --- 13 4 3 --- 1 1 --- --- St John’s wort, flowering top 17 15 14 3 Dandelion, root 5 2 1 --- Herbal drug Number of batches analysed Number of batches Cd > 0.5 mg/kg 5 Number of batches Cd > 1.0 mg/kg --- Water plantain, rhizome 2 1 1 --- Willow, bark 14 14 13 13 Raw materials tested included medicinal plants where cadmium could be commonly accumulated. Results showed that over a total of 113 different sample batches tested, corresponding to 21 different herbal drugs, 58 batches (51%) showed levels higher than 0.3 mg/kg, 45 batches (40%), levels over 0.5 mg/kg and 16 batches (14%), levels higher than 1 mg/kg (see Table 2). It was, moreover, observed that contamination with lead occurs rather by chance, whereas enhanced cadmium values are restricted to some species having a tendency to accumulate this heavy metal [13]. Some such species are St John’s wort [5,13,25,32], yarrow [5,13], linseed [15,22,27,32,33], German chamomile, absinth [13], valerian, passionflower, echinacea [25] and cinchona [34]. 1.2. Mercury: In the environment, several forms of mercury occur: elemental mercury (Hg(0)), monovalent mercury (Hg(I)) as mercurous chloride (HgCl), divalent mercury (Hg(II)) as mercuric chloride (HgCl2), and mercuric sulfide (HgS) or organic mercury, where it has formed compounds with carbon. Methylmercury (CH3Hg+), the most common example of this form of mercury, is particularly toxic (see II Health hazard). There are few anthropogenic sources of methylmercury pollution. Methylmercury is formed from divalent inorganic mercury by action of anaerobic sulfate- reducing bacteria that live in aquatic systems including lakes, rivers, sediments and the ocean. It is the form of mercury that is the most easily bioaccumulated in organisms. It is biomagnified in aquatic food chains from bacteria, to plankton, through macroinvertebrates, to herbivorous fish and to piscivorous (fish-eating) fish. The concentration of methylmercury in the top level aquatic predators can reach a level a million times higher than the level in water. When analyzed, mercury was never detected in samples of herbal drugs [23,26,35]. From 12,000 plant samples tested, it was concluded that its content was insignificant with regard to a threshold value of 0.1 mg/kg [14]. 1.3. Other elements: Very little information is available regarding the presence of other elements in herbal drugs and their preparations. Chromium and arsenic were undetectable above their limits of detection in different formulations prepared from ginseng [26]. In one study, where samples of herbal drugs were tested for thallium concentration, all 80 samples tested contained less than 0.01 mg/kg [15]. 1.4. Cadmium, mercury and arsenic in seaweed: Macroalgae can survive in toxic-metal-contaminated aquatic environments. In particular, Fucus spp. may accumulate heavy metals and have been widely used as biomonitors of metal pollution. So, they may be 2110 Natural Product Communications Vol. 3 (12) 2008 suitable species for use in risk assessment, for example for coast and estuarine areas. This tolerance to heavy metals can be explained in bladderwrack (Fucus vesiculosus L.) by its exceptional metalbinding properties due to metallothioneins, which are thought to act as a protective mechanism against incoming toxic metals, such as cadmium and mercury, [36,37]. Concentration of cadmium in bladderwrack has been extensively studied, showing that the level in seaweed varies with environmental levels [38-41]. Cadmium levels in bladderwrack reached values in the range of 15-25 mg/kg [39-41]. Concentrations of cadmium are highest in spring and lowest in autumn in F. vesiculosus and F. serratus [38,39]. Mercury levels in Fucus species (F. vesiculosus, F. spiralis and F. ceranoides) were found in the range 0.01-0.06 mg/kg for receptacles and 0.03-0.29 mg/kg for holdfast and stipe [42]. Levels of arsenic are higher in the aquatic environment than in most areas of land as it is fairly water-soluble and may be washed out of arsenicbearing rocks. In particular, seaweed is known to contain high concentrations of arsenic in comparison with terrestrial plants, owing to the ability of marine plants to concentrate the arsenic they derive from sea water. F. vesiculosus and Laminaria digitata contain levels of arsenic typically between 20 and 100 mg/kg dry weight [43,44]. Arsenic compounds can be categorized as inorganic (without an arsenic-carbon bond) and organic compounds (with an arseniccarbon-bond). Arsenic exists in three common oxidation states: As(0) (metalloid arsenic, 0 oxidation state), As(III) (trivalent state, such as arsenites), and As(V) (pentavalent state, such as arsenates). More than 90% of arsenic present in kelp is in the form of arsenosugars (arsenoribofuranosides), while inorganic arsenic is less than 5% [43]. Total and “reducible” arsenic levels (the latter level reflects the total of inorganic arsenic and some unstable organoarsenic species and is thus regarded as an indicator of maximum inorganic arsenic content) were analyzed in kelp food supplements on sale in the UK. They yielded mean concentrations of 25 mg/kg and 0.14 mg/kg for total and reducible arsenic, respectively. Potential daily intakes of reducible arsenic (based on maximum manufacturer’s dose recommendations) ranged from 0.05 to 9 µg [45]. Inorganic arsenic was found at concentrations in the range 67-96 mg/kg in hijiki seaweed [Hizikia fusiforme (Harv.) Okamura], a brown sea vegetable Guédon et al. growing wild on rocky coastlines around Japan and China [46]. The UK Food Standards Agency issued advice to consumers to avoid eating it [47]. 2. TOXIC METALS IN ASIAN TRADITIONAL MEDICINES Publications about unacceptable levels of toxic metals in non-Western folk medicines have regularly appeared in medical and pharmaceutical literature [48-53]. The presence of those poisonous metals is usually not due to contamination but to deliberate inclusion for alleged medicinal purpose or to an accidental addition in herbal preparations during their manufacture. In particular, Chinese and Indian preparations have been implicated. Taking into account that in most developed countries, Asian herbal medicines are becoming more and more popular [54], it seems justified to describe in this article those preparations which might constitute a serious health problem. Evidence suggesting that some Asian herbal medicines contain toxic metals have been reviewed [19,55]. For example, traditional Chinese medicines, which are usually complex mixtures of several (often 20 or more) herbal drugs, may contain heavy metals such as arsenic, cadmium, mercury and lead (see [48,50,51,56] for a review). Similarly, Indian medicinal systems (e.g. Ayurveda and Unani) have a long and rich history of herbal medicine, and heavy metals have been regular constituents of traditional Indian remedies (see [57] for a review). Surveys carried out during 2004 in the United States have shown that 20% of all Ayurvedic medicines in the Boston area contained potentially harmful levels of lead, mercury or arsenic [52]. Most common ingredients deliberately included for a specific curative purpose are realgar (arsenic sulfide), cinnabar (mercuric sulfide), calomel (mercurous chloride), hydrargyri oxydum rubrum (red mercuric oxide) and lithargyrum (lead monoxide) [58-60]. The presence of heavy metals may also be the result of accidental contamination during manufacture, for instance, from grinding weights [61] or lead-releasing containers [62] or other manufacturing utensils. 3. HEALTH HAZARD The main threats to human health from heavy metals are associated with exposure to lead, cadmium, mercury and arsenic. They have no known vital or beneficial effect on organisms, and their toxicity often takes years to manifest and may go unsuspected Toxic metals in herbal medicinal products Natural Product Communications Vol. 3 (12) 2008 2111 [63]. They have been extensively studied and their effects on human health regularly reviewed by international bodies, such as WHO (see [4] for a review). The formula, given in the section dealing with pesticide residues [3] for calculation of theoretical maximum tolerable levels in food commodities (mg/kg), from ADI values, can also be used to derive similar levels for a toxic metal [8]: Chronic exposure to cadmium can cause nephrotoxicity in humans, mainly due to abnormalities of tubular re-absorption [64]. Lead and mercury can cause adverse effects on the renal and nervous systems and can cross the placental barrier, with potential toxic effects on the fetus [65, 66]. The International Agency for Research on Cancer (IARC) classified cadmium and lead as human carcinogens: group I (sufficient evidence in both humans and experimental animals), and “possible human carcinogen”: group IIA (sufficient animal data and insufficient human data), respectively [67,68]. Longterm exposure to arsenic is mainly related to increased risks of skin, liver, bladder and lung cancer [69,70]. PTWI x M MWI x 100 M = body mass in kilograms (60 kg), MWI = mean weekly intake of raw material, in kilograms, 100 = general safety factor. PTWI values established by JECFA for cadmium [71], lead [72], mercury [71, 72] and arsenic [73] are listed in Table 3. The EU Scientific Committee on Food (SCF) adopted an opinion endorsing the PTWI established by JECFA (see [74]). 3.1. Mercury Trace elements (e.g. iron, cooper, nickel) may also have toxicity when considered in abnormally high doses. Metals in an oxidation state abnormal to the body may also become toxic: chromium (III) is an essential trace element, but chromium (VI) is a carcinogen. Radioactive metals (e.g. thorium, uranium, polonium) have both radiation toxicity and chemical toxicity. FAO/WHO recommended a PTWI value of 5 µg/kg/week for mercury, while the PTWI for methylmercury of 3.3 µg/kg/week [72] was revised by the 61st meeting of JECFA to 1.6 µg/kg/week [71]. Ingested methylmercury is readily and completely absorbed by the gastrointestinal tract. It is mostly found complexed with cysteine and with peptides and proteins containing that amino acid. The methylmercuric-cysteinyl complex is recognized by amino acid transporting proteins in the body, as methionine, another essential amino acid. Because of this mimicry, it is transferred freely throughout the body, including across the blood-brain barrier and across the placenta, where it is absorbed by the developing fetus. Because of this mimicry and its strong binding to proteins, methylmercury is not readily eliminated. Contrary to pesticides, where the toxicological risk is defined on the basis of Acceptable Daily Intake (ADI) values (see “Health hazard” under [3]), it is preferable for toxic metals to base the risk evaluation on the so-called Provisional Tolerable Weekly Intake (PTWI) values, established also by the Joint FAO/WHO Expert Committee on Food Additives (JECFA). PTWI is an endpoint used for food contaminants such as toxic metals with cumulative properties. This value (µg/kg body weight/week) represents permissible human weekly exposure to those contaminants unavoidably associated with the consumption of foods. 3.2. Arsenic: FAO/WHO has established a PTWI value only for inorganic arsenic (15 µg/kg/week) without assigning any such value to organic arsenicals [73]. JECFA are scheduled to Table 3: Provisional Tolerable Weekly Intake (PTWI) values for toxic metals, as established by the joint FAO/WHO Expert Committee on Food Additives (JECFA). Metal Lead Cadmium Mercury Arsenic (inorganic) PTWI (µg/kg/week) Reference 25 7 51 152 JECFA 2000 [72] JECFA 2003 [71] JECFA 2000 [72] JECFA 1989 [73] 1- But not more than 1.6 µg/kg/week in the form of methylmercury [71]. 2- This PTWI does not refer to organoarsenicals, as the organoarsenic compounds naturally occurring in marine products are considerably less toxic than inorganic arsenicals. 2112 Natural Product Communications Vol. 3 (12) 2008 review the PTWI for arsenic. The relative toxicity of an arsenical depends primarily on inorganic or organic form, oxidation state, solubility and rates of absorption and elimination. Inorganic arsenic is more toxic than organic arsenic (e.g. LD50 for arsenic trioxide in rats is 20 mg/kg, while for arsenobetaine no signs of toxicity were observed in mice after an oral dose of 10 g/kg) [75,76]. The toxicity of As(III) is several times greater than that of As(V), due to a greater cellular uptake. However, at equivalent intracellular levels, As(III) and As(V) compounds are equipotent. For instance, organoarsenic compounds, naturally occurring in fish (mainly arsenobetaine), are excreted very rapidly by man and there have been no reports of ill effects among populations whose consumption of large quantities of fish results in organoarsenical intakes of about 50 µg/kg per day [73]. Although the arsenic most commonly found in seaweed (organosugars) is relatively non-toxic to humans as compared with inorganic species [77], case reports of potential arsenic toxicosis secondary to kelp supplements intake have, surprisingly, been described [78,79]. 3.3. Other toxic metals: No FAO/WHO PTWI value for thallium has been established. The current dietary intake of this toxic element in the United Kingdom, which is estimated to be 5 µg/day, is not regarded as a cause for concern [80]. 3.4. Comments:The following points deserve special consideration: • The safety margin between a PTWI and the weekly exposure that produces deleterious effects can be relatively small. For instance, the critical organ in relation to the toxic effects of chronic ingestion of small amounts of cadmium is the kidney. So, intakes as low as 140-255 µg/day (PTWI = 60 µg/day/person with 60 kg bw) have been associated with low molecular weight proteinuria (without specific histological changes) in the elderly and renal dysfunction would be expected in sensitive population groups at cadmium exposure levels half that of the present PTWI, i.e. 30 µg/day [64]. On the other hand, a short-term exposure to levels exceeding the PTWI is not necessarily a cause for concern, provided the average intake over longer periods does not exceed the level set [73]. • As PTWI values refer to total dietary intakes, it is impossible to determine the acceptability of a certain contamination level in a herbal drug without considering the normal Guédon et al. dietary exposure to the metal in question. Dietary intakes measured in seven different countries [81] showed that the normal daily diet may provide a substantial portion of the tolerable daily amount that can be derived from the PTWI, dietary load of cadmium, lead and mercury accounting for 17-55%, 6-42% and 2-31% of tolerable daily amounts. Meanwhile, in many countries, lead intake from the diet can approach or exceed the PTWI [82]. Mercury exposure for the general population occurs mainly from consumption of fish [82] and possibly from dental amalgam fillings [66]. • Experts are still debating whether PTWI for cadmium can be used in general or whether it should be applied only to healthy adults, with the exclusion of risk groups such as chronically ill patients, pregnant women, and breast-feeding mothers [8]. Three other organizations, the National Institute of Public Health and the Environment (RIVM, the Netherlands), the Agency for Toxic Substances and Disease Registry (ATSDR, the United States) and the United States Environmental Protection Agency (US EPA) have also evaluated the noncancer oral toxicity data and established risk values for lead [83], cadmium [83,84], mercury (mercuric chloride, methylmercury) [83,85-87] and arsenic [83,88,89] (see Table 4). Critical organs or effects are: • brain and central nervous system (lead), • kidney damage (cadmium), • kidney, autoimmune effects (inorganic mercury), • central nervous system, developmental effects (methylmercury), • skin (inorganic arsenic). On the basis of sufficient evidence for an increased risk for cancer of the urinary bladder, lung and skin, an oral risk value has only been established for inorganic arsenic (6.7 ng/kg/day) [89]. 3.5 Toxic metals in Asian traditional medicines: The presence of heavy metals in ethnic medicines is considered as being a significant international problem. Lead and, less frequently, mercury and arsenic poisonings, associated with intentional or accidental addition of toxic heavy metals in unlicensed Ayurvedic medicines, have been described in developed countries with some regularity during the last three decades (see [57,90] for a review). Similarly, numerous case reports and Toxic metals in herbal medicinal products Natural Product Communications Vol. 3 (12) 2008 2113 Table 4: Other oral risk values for toxic metals. Metal Lead Cadmium Mercury (mercuric chloride) Methylmercury Arsenic (inorganic) Risk value1 RIVM [83] 3.6 µg/kg/day 25 µg/kg/week 0.5 µg/kg/day 3.5 µg/kg/week 2 µg/kg/day 14 µg/kg/week 0.1 µg/kg/day 0.7 µg/kg/week 1 µg/kg/day 7 µg/kg/week Risk value2 ATSDR [84, 85, 88] Risk value3 US EPA [86, 87, 89] ----- ----- 0.2 µg/kg/day 1.4 µg/kg/week ----0.3 µg/kg/day 2 µg/kg/week 0.3 µg/kg/day 2 µg/kg/week ----0.3 µg/kg/day 2 µg/kg/week 0.1 µg/kg/day 0.7 µg/kg/week 0.3 µg/kg/day 2 µg/kg/week 1- National Institute of Public Health and the Environment (RIVM): Tolerable Daily Intake (TDI). 2- Agency for Toxic Substances and Disease Registry (ATSDR): chronic oral Minimal Risk Level (MRL). 3- United States Environmental Protection Agency (US EPA): Reference Dose (RfD). case series of heavy metal poisoning associated with the use of traditional Chinese medicines (TCM) have been published in different countries (United States, Hong-Kong, Taiwan) (see [48] for a review). Recently, out of 247 TCM products investigated in Australia, some preparations exceeded FAO/WHO PTWI for arsenic (4 products), lead (1 product) and mercury (5 products), taking into account recommended product dose and the concentration of element in the product. The levels were high, ranging up to three orders of magnitude (103) higher than the PTWI for those elements considered [91]. It has also been shown that the passage of lead and cadmium into an extraction fluid decreased as the polarity of the extraction fluid diminished [7]. Thus, there can be little doubt that processing of herbal drugs with hydroethanolic mixtures likewise results in incomplete passage of heavy metals. 4. EFFECTS OF PROCESSING 5. ANALYTICAL METHODS The effects of extraction with boiling water were studied by analyzing lead and cadmium levels in 136 samples of 19 herbal drugs and in tisanes prepared from these samples. Passage into water was above 50% in only 12% and 8% of the lead and cadmium assays, respectively. The majority of tea samples (67% for lead and 71% for cadmium) showed a relatively low extraction of 25% or less. Individual extraction values ranged from 0.1% to 87% for lead and 1% to 68% for cadmium [21, 22]. Otherwise, the average percentage of lead and cadmium into the infusion from herbal teas consumed in Thailand was 6-12% and 14-24%, respectively [92] and determination of lead content in infusions or decoctions obtained from 11 herbal teas showed extraction values ranged from less than 1% to 59% [30]. Taking into account the relatively low concentrations of investigated toxic metals and complexity of the plant matrices, adequate physical techniques are necessary. Different methods have been recommended for quantitative analysis of plant samples, in order to satisfy relevant analytical criteria such as specificity and precision, and to provide less time consuming and cost beneficial results: • flame atomic absorption/emission spectrometry (FAAS/FAES); • graphite furnace atomic absorption spectrometry (GFAAS); • energy disperse X-ray fluorescence (EDX-RF); • inductively coupled plasma-atomic emission spectrometry (ICP-AES); • inductively coupled plasma-mass spectrometry (ICP-MS). When an herbal drug is contaminated at its surface with inorganic salts, it is likely that a relatively large amount will dissolve in the hot water. However, when metal traces are organically bound in the plant cell, the passage rate into a tisane will be relatively low [21, 22]. Besides, higher amounts of heavy metals are transferred from the raw material to water by boiling (decoction) than by immersion in hot water (infusion) [12]. Although FAAS/FAES and GFAAS are efficient to determine low levels of elements, techniques such as ICP-AES and mainly ICP-MS are superior due to multielement capabilities, low detection limits, isotopic capabilities and speed of analysis. The general chapter of the Ph. Eur. “Heavy metals in herbal drugs and fatty oils” (2.4.27) provides test methods for the estimation of lead, cadmium, mercury, arsenic, nickel, copper, iron and zinc by The washing of plants while they are still fresh may remove about 15-30% of heavy metal contamination [8]. For example, washing valerian roots after harvest has shown to be an effective way in reducing lead and cadmium contamination in crops [18]. 2114 Natural Product Communications Vol. 3 (12) 2008 atomic absorption spectroscopy (2.2.23). The content of arsenic and mercury is measured by direct calibration (Method I) using an automated continuous-flow hydride vapour generation system, while the content of other metals is determined by the standard additions method (Method II) using a graphite furnace as atomisation device. Other physical methods recently introduced in the 6th edition of the Ph. Eur. (6.0 January 2008) are ICPAES (2.2.57) and ICP-MS (2.2.58). The pharmacopoeial test for heavy metals (2.4.8), despite its limitations, is still a standard test described in individual monographs of the Ph. Eur. dealing mainly with chemical substances. Some improvements to simplify the test, avoiding loss of analytes and increasing sensitivity, have recently been proposed [93]. Methods used for the investigation of contaminants in the food area, i.e. standard procedures established by the European committee for standardization (CEN: Comité européen de normalisation), include determination of different trace elements. General aspects of analytical methodologies, sample preparation, quantitative analysis, precision of the method and results of collaborative tests are fully presented. Published standard procedures are listed below. 5.1. Cadmium and lead • Determination of lead, cadmium, zinc, copper, iron and chromium by atomic absorption spectrometry (AAS) after dry ashing (EN 14082: 2003) [94]; • Determination of lead, cadmium, chromium and molybdenum by graphite furnace atomic absorption spectrometry (GFAAS) after pressure digestion (EN 14083: 2003) [95]; • Determination of lead, cadmium, zinc, copper and iron by atomic absorption spectrometry (AAS) after microwave digestion (EN 14084: 2003) [96]. 5.2. Mercury • Determination of mercury by cold-vapour atomic absorption spectrometry (CVAAS) after pressure digestion (EN 13806: 2003) [97]. 5.3. Arsenic • Determination of total arsenic by hydride generation atomic absorption spectrometry Guédon et al. (HGAAS) after dry ashing (EN 14546: 2005) [98]; • Determination of total arsenic and selenium by hydride generation atomic absorption spectrometry (HGAAS) after pressure digestion (EN 14627: 2005) [99]; • Determination of inorganic arsenic in seaweed by hydride generation atomic absorption spectrometry (HGAAS) after acid extraction (EN 15517, 2008) [100]. 5.4. Cadmium, lead, mercury and arsenic • Determination of arsenic, cadmium, mercury and lead in foodstuffs by inductively coupled plasma-mass spectrometry (ICP-MS) after pressure digestion (standard project EN 15763: 2008) [101]. 6. STANDARDS/GUIDELINES 6.1. Foodstuffs: Commission Regulation 1881/2006/EC from December 2006 sets maximum levels for certain contaminants in foodstuffs, including maximum levels for toxic metals [74]. Maximum levels in vegetables, fruits, fresh herbs and fungi are in the range of 0.10-0.30 mg/kg and 0.050.20 mg/kg for lead and cadmium, respectively (see Table 5). Mercury is only controlled in fishery products, considering that levels of mercury found in foods other than fish and seafood are of lower concern. Moreover, it is mentioned that “the forms of mercury present in these other foods are mainly not methylmercury and they are therefore considered to be of lower risk” [74]. It should be noted that Commission Directive 61/2004/EC specifies a limit for mercury compounds expressed as mercury (pesticides prohibited for use in European Community) in food commodities of plant origin at the limit of analytical determination (0.01 mg/kg for food of plant origin and 0.02 mg/kg for tea and hops [102]. French regulation sets maximum levels for mineral arsenic, iodine and certain metals in seaweeds intended for human consumption [103]. Limits (values apply to the dried material) are as follows: • mineral arsenic : ≤ 3 mg/kg; • iodine : ≤ 5,000 mg/kg; • cadmium : ≤ 0.5 mg/kg; • mercury : ≤ 0.1 mg/kg; • lead : ≤ 5 mg/kg; • tin : ≤ 5 mg/kg. Toxic metals in herbal medicinal products Natural Product Communications Vol. 3 (12) 2008 2115 Table 5: Maximum limits of metals in foodstuffs (EU regulation). Foodstuffs Lead (mg/kg wet weight Cereals Vegetables Leaf vegetables, fresh herbs, cultivated fungi Fruit, excluding berries and small fruit Berries and small fruit Regulation 1881/2006/EC [74] Cadmium (mg/kg wet weight) 0.20 0.10 0.30 0.10 0.20 0.10 0.050 0.20 --------- Table 6: Maximum limits of metals in food supplements [104]. Heavy metals Foodstuffs Maximum levels (mg/kg wet weight) 3.0 Lead Food supplements1, dried seaweed1 Cadmium Food supplements1 1.0 Cadmium Food supplements consisting exclusively or mainly of dried seaweed or of products derived from seaweed, dried seaweed1 Food supplements1, dried seaweed1 3.0 Mercury 0.1 1- The maximum level applies to the finished product. 6.2. Food supplements 6.3. Herbal Medicinal Products It has been shown that food supplements can contribute significantly to human exposure to lead, cadmium and mercury as high levels of these toxic metals have been found in some food supplements during monitoring activities in European member states. In order to protect public health, the Commission of the European Communities considers that it is appropriate to set maximum levels for lead, cadmium and mercury in food supplements via an amendment of the Regulation 1881/2006/EC. A revised draft Commission Regulation has recently been released including the following comments and proposals [104]: • maximum levels must be safe and as low as reasonably achievable (ALARA), based upon good manufacturing practices. Proposals for limits are listed in Table 6; • due to natural accumulation of cadmium in seaweed, food supplements consisting of either dried seaweed or of products derived from seaweed can contain higher levels of cadmium than other food supplements. So, a higher maximum level for cadmium is needed for food supplements consisting exclusively or mainly of seaweed (Table 6). 6.3.1. Pharmacopoeias: Up to now, there are no binding criteria established by Ph. Eur. for toxic metals in herbal drugs. At present, with a few exceptions, no limit is prescribed in individual monographs for herbal drugs. Exceptions are: • kelp (Ph. Eur.: 1426): lead (≤ 5 mg/kg), cadmium (≤ 4 mg/kg), mercury (≤ 0.1 mg/kg) and total arsenic (≤ 90 mg/kg); • linseed (Ph. Eur.: 0095): cadmium (≤ 0.5 mg/kg); • red vine (Ph. fr.): copper (≤ 200 mg/kg) [106]. The draft Regulation favours the control at the final product level. Maximum levels for heavy metals in food supplements have been set out so far only in Belgium at the national level [105]. Limits in final products as sold are: • mercury : ≤ 0.2 mg/kg; • cadmium : ≤ 0.5 mg/kg; • lead : ≤ 1 mg/kg; • arsenic : ≤ 1 mg/kg. Presently, the general monograph on “Herbal drugs” (1433) states that a potential risk must be considered and, unless included in an individual monograph, limits may be required if justified. A draft proposal for a revision of this monograph has recently been published [107] including, amongst changes, the introduction of limits for certain heavy metals. These are as follows “unless otherwise stated in an individual monograph or unless otherwise justified and authorised: • cadmium : ≤ 0.5 mg/kg; • lead : ≤ 5 mg/kg; • mercury : ≤ 0.1 mg/kg. It is also specified that “if required by the relevant authority or by the nature or origin of the herbal drug, suitable limits for the contents of arsenic, copper, iron, nickel and zinc are defined”. Additionally, the general monograph on “Extracts” (0765) specifies “where applicable, as a result of analysis of herbal drug or animal matter used for production and in view of the production process, tests for microbiological quality, heavy metals, 2116 Natural Product Communications Vol. 3 (12) 2008 aflatoxins or pesticide residues in the extract may be required”. In relationship to the introduction of proposed limits for certain heavy metals in the monograph “Herbal drugs” (see above), a provision has been introduced in a revised draft of the monograph “Extracts” [108]. Herbal drugs used for extraction could exceed the limits set for these ones “provided the finished extract complies with the requirements concerning heavy metals in the general monograph Herbal drugs (1433)”. 6.3.2. European guidelines: The guideline on “Good Agricultural and Collection Practice” (GACP) [109] specifies that “medicinal plants should not be grown in soil contaminated with sludge, heavy metals, residues, plant protection products or other chemicals etc.”. According to the CPMP guideline (EMEA) on “Quality” [110]: “as a general rule, herbal substances must be tested, unless otherwise justified, for microbiological quality and for residues of pesticides and fumigation agents, toxic metals, likely contaminants and adulterants”. For herbal preparations, the guideline comments “if deemed necessary by analysis of the starting material, tests on microbiological quality, residues of pesticides, fumigation agents, solvents and toxic metals should be performed”. The other CPMP guideline (EMEA) on “Specifications” [111] stresses the point that “the need for inclusion of tests and acceptance criteria for inorganic impurities should be studied during development and based on knowledge of the plant species, its cultivation and the manufacturing process. Acceptance criteria will ultimately depend on safety considerations”. It is also mentioned regarding herbal preparations that “the potential for manufacturing process to concentrate toxic residues should be fully addressed. If the manufacturing process will reduce the burden of toxic residues, the tests on the herbal substance may be sufficient”. 6.3.3. National regulations: The German Ministry of Health published in 1991 a “draft recommendation for limits of heavy metals in medicinal products of plant and animal origin” [112]. This draft included the following limits for plants, parts of plants, oils, fats and waxes of plant origin and products thereof as well as for other products of plant origin, each with reference to the dried matter: • lead : ≤ 5 mg/kg; • cadmium : ≤ 0.2 mg/kg; • mercury : ≤ 0.1 mg/kg. Guédon et al. The following exceptions were considered for cadmium: 0.3 mg/kg for linseed, hawthorn, yarrow and 0.5 mg/kg for birch, St John’s wort, willow and maté. The draft recommendation has never been finally adopted. However, these maximum levels have been, so far, widely used as acceptance criteria for the assessment of marketing authorisation/registration application dossiers by EU national regulatory authorities. 6.4. WHO WHO guidelines on “Quality control methods for medicinal plants” [113] and “Quality of herbal medicines with reference to contaminants and residues” [114] recommend to take into consideration maximum limits of heavy metals. The following levels in dried plant material, based on ADI values, have been proposed: • lead : ≤ 10 mg/kg, • cadmium : ≤ 0.3 mg/kg. The following recommendation is given “the need for the inclusion of tests for toxic metals and acceptance criteria should be studied at the various development stages of the plant and based on knowledge of the medicinal plant species, its growth and/or cultivation and the manufacturing process” [114]. Additionally, if the heavy metals burden of the herbal material is unknown, “it is suggested that it can be determined qualitatively and quantitatively on several batches, preferably collected over several years” [114]. 7. LITERATURE RECOMMENDATIONS The German pharmaceutical manufacturers association (BAH), based on results collected from more than 20,000 samples by its contaminant working group (see also “I.1.1. Cadmium and lead”), showed that recommendations of the German Ministry of Health (see above “V.3.3. National regulations”) could not generally be applied [14, 115]. So, it made a proposal in 2002 for general limits: • lead : ≤ 10 mg/kg; • cadmium : ≤ 1 mg/kg; • mercury : ≤ 0.1 mg/kg; • arsenic : ≤ 5 mg/kg; • nickel : ≤ 10 mg/kg; • copper : ≤ 40 mg/kg. Based on an appropriate amount of data available, 90th percentile appears to be a practical limit in view of the limited availability of many herbal drugs. It is Toxic metals in herbal medicinal products Natural Product Communications Vol. 3 (12) 2008 2117 Table 7: Proposal of cadmium maximum levels for certain herbal drugs (BAH proposal) [116]. Cadmium : ≤ 0.3 mg/kg Wormwood Angelica (root) Arnica Hawthorn (leaf & flower) Elecampane Artichoke Milk thistle Damiana Strawberry Raspberry Gentian Kava-kava Linseed German chamomile Nettle (root) Passionflower Buckhorn plantain Silver linden (flower) Valerian Cadmium : ≤ 0.5 mg/kg Yarrow Cornflower Birch Buckthorn St Benedic thistle Coughgrass Mistletoe Mashmallow Rupturewort Immortelle Island moss Lovage Maté St John’s wort (herb) Lily of the valley Dandelion (root) Common horsetail Lungwort Roselle Wild thyme Pot marigold Stevia Speedwell Specific limits regarding cadmium : Belladonna (leaf) English holly Wild pansy Cinchona ≤ 1.0 mg/kg ≤ 3.5 mg/kg ≤ 1.5 mg/kg ≤ 1.5 mg/kg Willow Sanicle Thyme Zedoary ≤ 3.0 mg/Kg ≤ 0.6 mg/Kg ≤ 0.8 mg/Kg ≤ 1.0 mg/Kg considered that the usual small quantities of herbal drugs consumed do not pose a health hazard to the population [14]. As already described (see “I.1.1. Cadmium and lead”), many herbal drugs have a 90th percentile over 0.5 mg/kg (and even over 1.0 mg/kg) of cadmium [14]. So, it was observed that exceptions should be established for specific herbal drugs with 90th percentile higher than 1 mg/kg, e.g. St John’s wort, willow and fucus. Previously, based on a restrictive cadmium maximum level of 0.2 mg/kg [112], BAH made, in 1992, a proposal for exemptions of a large number of herbal drugs known to have a specific affinity to cadmium [116] (see Table 7). 8. CONCLUDING REMARKS Contamination of medicinal plants and herbal drugs by heavy metals has been extensively documented in published literature [5-16,18,20-35]. Based on the present survey, different observations can be made: • first of all, the deliberate addition or not (accidental contaminations during manufacturing) of compounds such as cinnabar (HgS) or realgar (As4S4) to certain preparations, allowable by Chinese and Indian traditional medicines [19,48-62,90,91], must be clearly differentiated from the presence of heavy metals due to environmental pollution. Such unacceptable levels of toxic metals in non- Western traditional herbal remedies should not overestimate the real risk due to the presence of toxic metals in herbal drugs; • mercury: no literature has been found supporting a risk of level above a threshold value of 0.1 mg/kg in medicinal plants [14, 23, 26, 35], with the exception of seaweed like bladderwrack where it may accumulate [42]. Toxicity is mainly due to methylmercury as it is readily and completely absorbed by the gastrointestinal tract. Mercury exposure and especially its organic form, methylmercury, for the general population occurs mainly from consumption of fish and seafood [82], which explains why maximum levels in foodstuffs apply only to fishery products [74]; • arsenic: the ability to concentrate arsenic has been described in marine plants such as kelp [4345]. Organic arsenic is much less toxic than inorganic arsenic (e.g. arsenosugars) due to its rapid excretion by man, thereby a PTWI value has only been established for inorganic arsenic [73]. The three main factors which may lead to high levels of toxic metals in medicinal plants are the following: • anthropogenic influence: human activities lead to the production of available (soluble) forms of toxic metals in the soils, the main source of such elements for terrestrial plants. Furthermore, some anthropogenic activities may also cause heavy metals contamination of plants by deposition of air particles on the aboveground organs. Contamination via air pollution may be the most significant source of heavy metals in certain areas (e.g. vicinity of mining industry, smelting works, waste incineration plants). Agricultural chemicals may also contribute to high levels of toxic metals (e.g. phosphate fertilizers, which contain varying quantities of cadmium depending on the original content of the phosphate rocks, sewage sludge-amended soils, pesticides, such as lead arsenate or mercury-containing products which continue to be used today in some countries) [4,6,810,15,16,18,114]; • availability in the soil: soil characteristics such as pH, redox-potential, content and form of organic matter or clay minerals affect heavy metal chemical behaviour in soils. Amongst these factors, soil pH is probably the most important one (availability of all elements, with the 2118 Natural Product Communications Vol. 3 (12) 2008 exception of molybdenum, increases as pH decreases) [4,5]; • uptake by the plant: genetic features of certain plant species show a tendency to accumulate trace elements, the most characteristic being cadmium [6,12,14]. Plants which are prone to increase assimilation of that element are named “cadmium collector” (e.g. St John’s wort, willow, yarrow, linseed, birch, wild pansy, dandelion) [5,13,14,22,25,27,31,32]. So, excluding species which have a natural tendency to accumulate cadmium, cultivated plant materials may be considered as being more at risk of contamination than raw materials collected in wild habitats. 9. GENERAL RECOMMENDATIONS As already mentioned, the CPMP guideline (EMEA) on “Specifications” [111] stresses the point that “the need for inclusion of tests and acceptance criteria for inorganic impurities should be studied during development and based on knowledge of the plant species, its cultivation and the manufacturing process”. Taking into account previous comments, criteria, which should be part of a risk assessment, include: • presence or not of anthropogenic influences in the vicinity of the site of cultivation or wild collection resulting in a potentially contaminated environment; • soil pH where the herbal drug is harvested/collected. Medicinal plants, as all plants, may exhibit or not different soil preferences, e.g. preference for soils rich or poor in lime (high and low pH values, respectively); • cultivation techniques (e.g. use of fertilizers); • ability of the plant species to uptake and accumulate toxic metals (mainly cadmium). Information justifying that testing heavy metals on a routine basis can be waived (i.e. no contamination to be expected following the risk assessment) should preferably be supported by development data, including at least three suitable representative batches. Assurance must also be provided that the medicinal plant was cultivated/collected in accordance with the principles of Good Agricultural and Collection Practice [109]. Moreover, heavy metals are most often weakly extracted into the herbal drug preparations during Guédon et al. processing (e.g. infusion, alcoholic extracts). Therefore, it is fully appropriate, as included in the revision draft for the Ph. Eur. monograph “Extracts” [107], to consider the possibility of using raw materials exceeding the limits set for herbal drugs (see proposed revision for the monograph “Herbal drugs” [108]), provided the herbal preparation complies with the herbal drugs requirements. 9.1. Cadmium and lead: Harmonised maximum limits which are scheduled in the revised pharmacopoeial monograph “Herbal drugs” (Ph. Eur. 1433) for cadmium (≤ 0.5 mg/kg) and lead (≤ 5 mg/kg) [107] are achievable for most herbal drugs. Meanwhile, exceptions for cadmium collectors need to be considered, some herbal drugs containing often cadmium levels above 0.5 mg/kg. It is presently the case for kelp where a maximum level of 4 mg/kg is set up (Ph. Eur.: monograph “Kelp” 1426). Assuming a mean daily consumption of 5 g herbal drug (average daily dose according to the monograph “Tisanes” of the French Pharmacopoeia [117]), a passage rate of heavy metals into hot water of 20% [21,22] and a concentration of 5 mg/kg lead and 0.5 mg/kg cadmium, the estimated weekly intake would represent 2% and 0.8% respectively of the PTWI recommended by the WHO for an adult of 70 kg (1,750 µg of lead and 490 µg of cadmium) [71,72]. So, the consumption of herbal drugs is not expected to contribute significantly to the exposure of the population to cadmium and lead. 9.2. Mercury: As mercury exposure through consumption of herbal drugs of terrestrial origin does not appear to be of health concern, as its uptake is insignificant, inclusion of the test is not justified. 9.3. Arsenic: Arsenic is not identified as being a risk in herbal drugs of terrestrial origin. Based on safety considerations, it is advisable, if necessary, to include in individual monographs testing of inorganic arsenic rather than total arsenic, based on standard EN 15517 [100]. The official monograph “Kelp” (Ph. Eur.: 1426) should be modified accordingly as only inorganic arsenic is to be considered a risk (presently, Ph. Eur. sets a limit of 90 mg/kg total arsenic in kelp). 9.4. Other trace elements: Only, if required by the nature or origin of the herbal drug, a suitable limit for the content of other trace elements (e.g. copper when used as fungicide) may be defined. Toxic metals in herbal medicinal products Natural Product Communications Vol. 3 (12) 2008 2119 9.5. Food supplements: Current proposals for legislation on heavy metals in food supplements [104] are not identical to those scheduled by the Ph. Eur. [108], i.e. for cadmium 1 mg/kg and 0.5 mg/kg, and for lead 3 mg/kg and 5 mg/kg, respectively. An harmonisation of maximum limits should be justified as food supplements and herbal remedies have similar specificities, i.e. they have a defined serving size (e.g. 1 tablet/day) with a small amount of product consumed daily in comparison to typical food and a duration of use which is also limited. Maximum levels scheduled by the Ph. Eur. [107] represent practicable limits on the basis of the present literature survey. Additionally, the contaminant limits should be set for the raw materials and not for the final products as sold, as specified in the draft Regulation [104]. References [1] [2] [3] [4] [5] [6] [7] [8] [9] [10] [11] [12] [13] [14] [15] [16] [17] [18] [19] [20] [21] [22] Commission SFSTP, Guedon D, Brum M, Bizet D, Bizot S, Bourny E, Compagnon PA, Kergosien H, Quintelas LG, Respaud J, Saperas O, Seigneuret JM, Taoubi K, Urizzi P. (2007) Impurities in herbal substances, herbal preparations and herbal medicinal products, I. Microbial contamination. STP Pharma Pratiques, 17, 183-208. Commission SFSTP, Guédon D, Brum M, Taoubi K, Bizet D, Bizot S, Bourny E, Compagnon PA, Kergosien H, Quintelas LG, Respaud J, Saperas O, Seigneuret JM, Urizzi P. (2007) Impurities in herbal substances, herbal preparations and herbal medicinal products, II. Mycotoxins. STP Pharma Pratiques, 17, 209-225. Commission SFSTP, Guédon D, Brum M, Saperas O, Quintelas LG, Bizet D, Bizot S, Bourny E, Compagnon PA, Kergosien H, Respaud J, Seigneuret JM, Taoubi K, Urizzi P. (2008) Impurities in herbal substances, herbal preparations and herbal medicinal products, III. Pesticide residues. STP Pharma Pratiques, 18, 111-136. Järup L. (2003) Hazards of heavy metal contamination. British Medical Bulletin, 68, 167-182. Radanovic D, Antic-Mladenovic S, Jakovljevic M. (2002) Influence of some soil characteristics on heavy metal content in Hypericum perforatum L. and Achillea millefolium L. Acta Horticulturae, 576, 295-301. Angelova V, Ivanov K, Ivanova R. (2005) Heavy metal content in plants from family Lamiaceae cultivated in an industrially polluted region. Journal of Herbs Spices and Medicinal Plants, 11, 37-46. Peters H, Schilcher H. (1986) Schwermetallbelastung von Arzneidrogen und deren Zubereitungen. Planta Medica, 52, 521-522. Schilcher H, Peters H, Wank H. (1987) Pestizide und Schwermettalle in Arzneipflanzen und Arzneipflanzen-Zubereitungen. Pharmazeutische Industrie, 49, 203-211. Little P, Martin MH. (1972) A survey of zinc, lead and cadmium in soil and natural vegetation around a smelting complex. Environmental Pollution, 3, 241-254. Wang QR, Cui YS, Liu XM, Dong YT, (2003) Christie P. Soil contamination and plant uptake of heavy metals at polluted sites in China. Journal of Environmental Science and Health, A38, 823-838. Adriano DC (1986) Trace elements in terrestrial environment. Springer-Verlag, New York. Abou-Arab AAK, Abou Donia MA. (2000) Heavy metals in Egyptian spices and medicinal plants and the effect of processing on their levels. Journal of Agricultural Food and Chemistry, 48, 2300-2304. Chizzola R, Michitsch H, Franz C. (2003) Monitoring of metallic micronutrients and heavy metals in herbs, spices and medicinal plants from Austria. European Food Research and Technology, 216, 407-411. Kabelitz L, Barbin Y. (1998) Les métaux lourds dans les plantes médicinales. STP Pharma Pratiques 1999;9:443-53 (KABELITZ L. Heavy metals in herbal drugs. European Journal of Herbal Medicine, 4, 25-33. Ali SL. (1983) Bestimmung der Pestiziden Rückstände und anderer bedenklicher Verunreinigungen– wie toxische Metallspuren –in Arzneipflanzen. 2. Mitteilung. Bestimmung von toxischen Metallspuren in Arzneidrogen. Pharmazeutische Industrie, 45, 1294-1295. Gosselin RE, Smith RP, Hodge HC, Braddock JE (1984) Clinical toxicology of commercial products, 5th ed. Williams & Wilkins, Baltimore. CSTEE (Scientific Committee on Ecotoxicity and the Environment). (2002) Opinion on “Member State assessments of the risk to health and the environment from cadmium in fertilizers”. Brussels. http://ec.europa.eu/health/ph_risk/committees/04_scher/docs/scher_o_043.pdf. Weightman RM. (2006) Heavy metal and microbial contamination of valerian (Valeriana officinalis L.) roots grown in soil treated with sewage sludge. Journal of Herbs Spices and Medicinal Plants, 12, 77-88. Ernst E. (2002) Toxic heavy metals and undeclared drugs in Asian herbal medicines. Trends in Pharmacological Sciences, 23, 136-139. Ali SL, Blume H. (1983) Bestimmung von Pestizid-Rückständen und toxischen Schwermetallspuren in Arzneidrogen. Pharmazeutische Zeitung, 128, 2248-2252. Ali SL. (1985) Bestimmung der Pestizidrückstände und toxischen Metallspuren in Teeaufgüssen aus Arzneidrogen. Pharmazeutische Zeitung, 130, 1927-1932. Ali SL. (1987) Bestimmung der Pestizidrückstände und toxischen Schwermetallspuren in Arzneidrogen und deren Teeaufgüssen. Pharmazeutische Zeitung, 132, 633-638. 2120 Natural Product Communications Vol. 3 (12) 2008 [23] [24] [25] [26] [27] [28] [29] [30] [31] [32] [33] [34] [35] [36] [37] [38] [39] [40] [41] [42] [43] [44] [45] [46] [47] [48] [49] [50] [51] [52] [53] [54] [55] [56] [57] Guédon et al. Caldas ED, Machado LL. (2004) Cadmium, mercury and lead in medicinal herbs in Brazil. Food and Chemical Toxicology, 42, 599-603. Dogheim SM, EL Ashraf MM, Alla SAG, Khorshid MA, Fahmy SM. (2004) Pesticides and heavy metals levels in Egyptian leafy vegetables and some aromatic medicinal plants. Food Additives and Contaminants, 21, 323-330. Hugget DB, Khan IA, Allgood JC, Block DS, Schlenk D. (2001) Organochlorine pesticides and metals in select botanical dietary supplements. Bulletin of Environmental Contamination and Toxicology, 66, 150-155. Khan IA, Allgood J, Walker LA, Abourashed EA, Schlenk D, Benson WH. (2001) Determination of heavy metals and pesticides in ginseng products. Journal of AOAC International, 84, 936-939. Schilcher H. (1982) Rückstände und Verunreinigungen bei Drogen und Drogenzubereitungen. 19. Mitteilung: Zur Wertbestimmung und Qualitätsprüfung von Drogen. Planta Medica, 44, 65-77. Schilcher H. (1985) Rückstandsbelastung von Drogen und Drogenzubereitungen. Deutsche Apotheker Zeitung, 125, 47-52. Schilcher H, Peters H. (1990) Empfehlung von Richt-und Grenzwerten für den maximalen Blei- und Cadmium-Gehalt von Arzneidrogen und daraus hergestellter pharmazeutischer Zubereitungen. Pharmazeutische Industrie, 52, 916-921. Slaveska R, Spirevska I, Stafilov T, Ristov T. (1998) The content of trace metals in some herbal teas and their aqueous extracts. Acta Pharmaceutica, 48, 201-209. EDQM/Official Medicines Control Laboratories: (2006) Market surveillance study 031. Determination of cadmium in herbal drugs. Schneider M, Marquard R. (1996) Aufnahme und Akkumulation von Cadmium und Schwermetalle bei Hypericum perforatum L. and Linum usitatissimum L. Arznei- Gewürzpflanzen, 1, 111-116. Anonymous. (1988) Zuviel Cadmium in Leinsamen. Deutsche Apotheker Zeitung, 128, 145-146. De Pasquale A, Paino E, De Pasquale R, Germano MP. (1994) Contamination by heavy metals in drugs from different commercial sources. Pharmeuropa, 6, 44-47. Raman P, Patino LC, Nair MG. (2004) Evaluation of metal and microbial contamination in botanical supplements. Journal of Agricultural Food Chemistry, 52, 7822-7827. Merrifield ME, Chaseley J, Kille P, Stillman MJ. (2006) Determination of the Cd/S cluster stoichiometry in Fucus vesiculosus metallothionein. Chemical Research in Toxicology, 19, 365-375. Morris CA, Nicolaus B, Sampson V, Harwood JL, Kille P. (1999) Identification and characterization of a recombinant metallothionein protein from a marine alga, Fucus vesiculosus. Biochemical Journal, 338, 553-560. Fuge R, James KH. (1973) Trace metal concentrations in brown seaweeds, Cardigan Bay, Wales. Marine Chemistry, 1, 281-293. Fuge R, James KH. (1974) Trace metal concentrations in Fucus from the Bristol Channel. Marine Pollution Bulletin, 5, 9-12. Morris AW, Bale AJ. (1975) The accumulation of cadmium, copper, manganese and zinc by Fucus vesiculosus in the Bristol channel. Estuarine and Coastal Marine Science, 3, 153-163. Söderlund S, Forsberg A, Pedersen M. (1988) Concentration of cadmium and other metals in Fucus vesiculosus L. and Fontinalis dalecarlica Br. Eur. from the northern Baltic Sea and the southern Bothnian Sea. Environmental Pollution, 51, 197-212. Cairrao E, Pereira MJ, Pastorinho MR, Morgado F, Soares AM, Guilhermo L. (2007) Fucus spp. as a mercury contamination bioindicator in coastal areas (Northwestern Portugal). Bulletin of Environmental Contamination and Toxicology, 79, 388-395. Castelhouse H, Smith C, Raab A, Deacon C, Meharg A, Feldmann J. (2003) Biotransformation and accumulation of arsenic in soil amended with seaweed. Environmental Science and Technology, 37, 951-957. Francesconi KA, Edmonds JS. (1997) Arsenic and marine organisms. Advances in Inorganic Chemistry, 44, 147-189. Norman JA, Pickford CJ, Sanders TW, Waller M. (1987) Human intake of arsenic and iodine from seaweed-based food supplements and health foods available in the UK. Food Additives and Contaminants, 5, 103-109. Rose M, Lewis J, Langford N, Baxter M, Origgi S, Barber M, Macbain H, Thomas K. (2007) Arsenic in seaweed– Forms, concentration and dietary exposure. Food and Chemical Toxicology, 45, 1263-1267. FSA. (2004) Food Safety Agency. Committee on toxicity of chemicals in food, consumer products and the environment. United Kingdom. http://www.food.gov.uk/multimedia/pdfs/TOX-2004-35.pdf. Ernst E, Coon TJ. (2001) Heavy metals in traditional Chinese medicines: a systematic review. Clinical Pharmacology and Therapeutics, 70, 497-504. Espinoza EO, Mann MJ. (1995) Arsenic and mercury traditional Chinese herbal balls. The New England Journal of Medicine, 333, 803-804. Ko RJ. (1998) Adulterants in Asian patent medicines. The New England Journal of Medicine, 339, 847. Koh HL, Woo SO. (2000) Chinese proprietary medicines in Singapore. Regulatory control of toxic heavy metals and undeclared drugs. Drug Safety, 23, 351-362. Saper RB, Kales SN, Paquin J, Burns MJ, Eisenberg DM, Davis RB, Phillips RS. (2004) Heavy metal content of Ayurvedic herbal medicine products. JAMA, 292, 2868-2873. Wong MK, Tan P, Wee YC. (1993) Heavy metals in some Chinese herbal plants. Biological Trace Element Research, 36, 135-142. Ernst E, White AR. (2000) The BBC survey of complementary medicine use in the UK. Complementary Therapies in Medicine, 8, 32-36. Ernst E. Harmless herbs? (1998) A review of the recent literature. The American Journal of Medicine, 104, 170-177. Chan K. (2003) Some aspects of toxic contaminants in herbal medicines. Chemosphere, 52, 1361-1371. Ernst E. (2002) Heavy metals in traditional Indian remedies. European Journal of Clinical Pharmacology, 57, 891-896. Toxic metals in herbal medicinal products [58] [59] [60] [61] [62] [63] [64] [65] [66] [67] [68] [69] [70] [71] [72] [73] [74] [75] [76] [77] [78] [79] [80] [81] [82] [83] [84] [85] [86] [87] [88] [89] [90] Natural Product Communications Vol. 3 (12) 2008 2121 Chan TYK, Tomlinson B, Critchley AJH. (1993) Chinese herbal medicines revisited: a Hong Kong perspective. The Lancet, 342, 1532-1534. Levitt C, Godes J, Eberhardt M, Ing R, Simpson JM. (1984) Sources of lead poisoning. JAMA, 252, 3127-3128. Yeung H-C. (1985) Handbook of Chinese herbs and formulas, vol II. Institute of Chinese Medicine, Los Angeles, CA. Yu ECL, Yeung CY. (1987) Lead encephalopathy due to herbal medicine. Chinese Medical Journal, 100, 915-917. Chia BL, Leng CK, Hsii FP, Yap MHL, Lee YK. (1973) Lead poisoning from contaminated opium. BMJ, 1, 354. Goyer RA, Klaassen CD, Waalkes MP. (1995) Metal toxicology. Academic Press, San Diego, CA. Nordberg G. (1999) Excursions of intake above ADI: Case study on cadmium. Regulatory Toxicology and Pharmacology, 30, S57-S62. Tong S, Von Schirnding YE, Prapamontol T. (2000) Environmental lead exposure: a public problem of global dimension. Bulletin of the World Health Organization, 78, 1068-1077. WHO. (2003) Elemental mercury and inorganic mercury compounds: human health aspects. Concise international chemical assessment document 50. World Health Organisation, Geneva. IARC Monographs. (1993) Cadmium and cadmium compounds. In: Beryllium, cadmium, mercury and exposure in the glass manufacturing industry, vol 58. Lyon: International Agency for Research on Cancer, 119-237. Steenland K, Boffetta P. (2000) Lead and cancer in humans: where are we now? American Journal of Industrial Medicine, 3, 295-299. WHO. (2001) Arsenic and arsenic compounds. Environmental health criteria, vol. 224. World Health Organisation, Geneva. IARC Monographs. (2002) Some drinking-water disinfectants and contaminants, including arsenic, vol 84. Lyon: International Agency for Research on Cancer. Summaries and evaluations http://monographs.iarc.fr/htdocs/announcements/vol84.htm. JECFA. (2003) Joint FAO/WHO Expert Committee on Food Additives. 61st meeting. Summary and conclusions. World Health Organization, Geneva. http://www.who.int/ipcs/food/jecfa/summaries/en/summary_61.pdf. JECFA. (2000) Joint FAO/WHO Expert Committee on Food Additives. 25th meeting. Summary and conclusions. World Health Organization, Geneva. JECFA. (1989) Joint FAO/WHO Expert Committee on Food Additives. Evaluation of certain food additives and contaminants. 33rd report. Technical Report Series 776. World Health Organization, Geneva. Commission Regulation 1881/2006/EC of 19 December 2006 setting maximum levels for certain contaminants in foodstuffs. Official Journal of the European Communities 2006:L364/5-24 (20 December 2006). WHO. (1981) Environmental health criteria 18, Arsenic. World Health Organisation, Geneva. Phillips DJ, Depledge MH. (1985) Metabolic pathways involving arsenic in marine organisms a unifying hypothesis. Marine Environmental Research, 17, 1-12. Sakurai T, Kaise T, Ochi T, Saitoh T, Matsubara C. (1997) Study of in vitro cytotoxicity of a water soluble organic arsenic compound, arsenosugar, in seaweed. Toxicology, 122, 205-212. Amster E, Tiwary A, Schenker MB. (2007) Case report: potential arsenic toxicosis secondary to herbal kelp supplement. Environmental Health Perspectives, 115, 606-608. Conz PA, La Greca G, Benedetti P, Bevilaqua PA, Cima L. (1988) Fucus vesiculosus: a nephrotoxic alga? Nephrology Dialysis Transplantation, 13, 526-527. Sherlock JC, Smart GA. (1986) Thallium in foods and the diet. Food Additives and Contaminants, 3, 363-370. Van Dokkum W, De Vos RH, Muys T, Wesstra JA. (1989) Minerals and trace elements in total diets in The Netherlands. British Journal of Nutrition, 61, 7-15. Baht RV, Moy GG. (1997) Monitoring and assessment of dietary exposure to chemical contaminants. Rapport Trimestriel Statistique Sanitaire Mondial, 50, 132-148. Baars AJ, Theelen RMC, Janssen PJCM, Hesse JM, Van ApeldoorN ME, Meijerink MCM, Verdam L, Zeilmaker MJ. (2001) Reevaluation of human-toxicological maximum permissible risk levels. RIVM report no. 711701025, National Institute of Public Health and the Environment, Bilthoven, The Netherlands,. Available at http://www.rivm.nl/bibliotheek/rapporten/711701025.pdf. ATSDR (Agency for Toxic Substances and Disease Registry). (1999) Toxicological profile for cadmium. U.S. Department of Health and Human Services, Public Health Service. Available at http://www.atsdr.cdc.gov/toxprofiles/tp5.html. ATSDR (Agency for Toxic Substances and Disease Registry). (1999) Toxicological profile for mercury. Update. U.S. Department of Health and Human Services, Public Health Service. Available at http://www.atsdr.cdc.gov/toxprofiles/tp46.html. U.S. EPA. (1995) Integrated Risk Information System (IRIS). Mercuric chloride (CASRN 7487-94-7). Online. National Center for Environmental Assessment, Washington, DC. Available at http://www.epa.gov/iris/subst/0692.htm. U.S. EPA. (2001) Integrated Risk Information System (IRIS). Methylmercury (CASRN 22967-92-6). Online. National Center for Environmental Assessment, Washington, DC. Available on http://www.epa.gov/iris/subst/0073.htm. ATSDR (Agency for Toxic Substances and Disease Registry). (2007) Toxicological profile for arsenic. U.S. Department of Health and Human Services, Public Health Service. Available at http://www.atsdr.cdc.gov/toxprofiles/tp2.html. U.S. EPA. (1998) Integrated Risk Information System (IRIS). Arsenic, inorganic (CASRN 7440-38-2). Online. National Center for Environmental Assessment, Washington, DC. Available at http://www.epa.gov/iris/subst/0278.htm. Shaw D. (1997) Traditional remedies and food supplements: a 5-year toxicological study (1991-1995). Drug Safety, 17, 342-356. 2122 Natural Product Communications Vol. 3 (12) 2008 [91] [92] [93] [94] [95] [96] [97] [98] [99] [100] [101] [102] [103] [104] [105] [106] [107] [108] [109] [110] [111] [112] [113] [114] [115] [116] [117] Guédon et al. Cooper K, Noller B, Connel D, Yu J, Sadler R, Olszowy H, Golding G, Tinggi U, Moore MR, Myers S. (2007) Public health risks from heavy metals and metalloids present in traditional Chinese medicines. Journal of Toxicology and Environmental Health A, 70, 1694-1699. Nookabkaew S, Rangkadilok N, Satayavivad J. (2006) Determination of trace elements in herbal tea products and their infusions consumed in Thailand. Journal of Agricultural Food Chemistry, 54, 6939-6944. Lodi A, Miller JHMB, Concessa JY, Nap CJ. (2007) The evolution of the pharmacopoeial test for heavy metals. Pharmeuropa Scientific Notes, 1, 33-37. EN 14082. (2003) Foodstuffs - Determination of trace elements - Determination of lead, cadmium, zinc, copper, iron and chromium by atomic absorption spectrometry (AAS) after dry ashing. EN 14083. (2003) Foodstuffs - Determination of trace elements - Determination of lead, cadmium, chromium and molybdenum by graphite furnace atomic absorption spectrometry (GFAAS) after pressure digestion. EN 14084. (2003) Foodstuffs - Determination of trace elements - Determination of lead, cadmium, zinc, copper and iron by atomic absorption spectrometry (AAS) after microwave digestion. EN 13806. (2003) Foodstuffs - Determination of trace elements - Determination of mercury by cold-vapour atomic absorption spectrometry (CVAAS) after pressure digestion. EN 14546. (2005) Foodstuffs - Determination of trace elements - Determination of total arsenic by hydride generation atomic absorption spectrometry (HGAAS) after dry ashing. EN 14627. (2005) Foodstuffs - Determination of trace elements - Determination of total arsenic and selenium by hydride generation atomic absorption spectrometry (HGAAS) after pressure digestion. EN 15517. (2008) Foodstuffs - Determination of trace elements - Determination of inorganic arsenic in seaweed by hydride generation atomic absorption spectrometry (HGAAS) after acid extraction. prEN 15763. (2008) Foodstuffs - Determination of trace elements - Determination of arsenic, cadmium, mercury and lead in foodstuffs by inductively coupled plasma mass spectrometry (ICP-MS) after pressure digestion. Commission Directive 61/2004/EC of 26 April 2004 amending the annexes to Council Directives 86/362/EEC, 86/363/EEC and 90/642/EEC as regards maximum residue levels for certain pesticides prohibited for use in the European Community. Official Journal of the European Union N° L 127/81-91 of 29 April 2004. LAMY-DEHOVE. (2002) Réglementation des produits, qualité, répression des fraudes. Etude 268-425 : critères toxicologiques exigés pour l’utilisation des algues en alimentation humaine, Lamy SA, Paris. SANCO/1524/2007 rev. 5. Draft Commission Regulation amending Regulation (EC) N° 1881/2006 setting maximum levels for certain contaminants in foodstuffs as regards heavy metals. http://ec.europa.eu/food/committees/regulatory/scfcah/toxic/ag_11042008_en.pdf. Arrêté royal fixant les teneurs maximales en contaminants comme les métaux lourds dans les compléments alimentaires (14 juin 2002). Moniteur Belge du 25 juillet 2002, p. 33138-9. Vigne rouge – Vitis vinifera (janvier 1996). Pharmacopée française Xe édition. Agence française de sécurité sanitaire des produits de santé. Herbal drugs. (2008) Pharmeuropa, 20, 302-303. Extracts. (2008) Pharmeuropa, 20, 291-293. EMEA/HMPC Guideline on good agricultural and collection practice (GACP) for starting materials of herbal origin (EMEA/HMPC/246816/05). EMEA Guideline on quality of herbal medicinal products/traditional herbal medicinal products (CPMP/QWP/2819/00 Rev 1). EMEA Guideline on specifications: tests procedures and acceptance criteria for herbal substances, herbal preparations and herbal medicinal products/traditional herbal medicinal products (CPMP/QWP/2820/00 Rev 1). Bundesministerium für Gesundheit. Bekanntmachung von Empfehlungen für Höchstmengen an Schwermetallen bei Arzneimitteln pflanzlicher und tierischer Herkunft (Arzneimittel-Kontaminanten-Empfehlungen – Schwermetalle). Entwurf vom 17.10.1991. WHO. (1998) Quality control methods for medicinal plant materials. World Health Organisation, Geneva. WHO. (2007) WHO guidelines for assessing quality of herbal medicines with reference to contaminants and residues. World Health Organisation, Geneva. Kabelitz L. (2003) Contamination of herbal drugs in regulation and future developments. Herba Polonica, 49, 89-104. Stellungnahme des Bundesfachverbandes der Arzneimittel-Hersteller e.V. (BAH) vom 14 Februar 1992 zum Entwurf des Bundesministeriums für Gesundheit vom 17.10.1991 “Bekanntmachung von Empfehlungen für Höchstmengen an Schwermetallen bei Arzneimitteln pflanzlicher und tierischer Herkunft (Arzneimittel-Kontaminanten-Empfehlungen – Schwermetale) ”. Tisanes. (2000) In Pharmacopée française Xe édition. Agence française de sécurité sanitaire des produits de santé. NPC Natural Product Communications A Fresh Insight into the Interaction of Natural Products with Pregnane X Receptor 2008 Vol. 3 No. 12 2123 - 2128 Salvador Máñez Departament de Farmacologia, Universitat de València, Av. Vicent Andrés Estellés s/n, 46100 Burjassot, Spain salvador.manez@uv.es Received: July 29th, 2008; Accepted: October 20th, 2008 The discovery that various drugs (e.g., phenobarbital) stimulate their own metabolism through a mechanism coined as enzymatic induction opened up a fascinating road that eventually led to the accurate biochemical characterization of the pregnane X receptor. After numerous studies, researchers have concluded that this receptor is activated by different endogenous steroids and a number of foreign lipophile ligands. Once activated, it induces the synthesis of oxygenases and conjugating enzymes. The activating ligands identified to date include many synthetic drugs, along with a number of natural products. The present review summarizes the data relating to the origin, chemistry, and pharmacological activity of the newest natural products that have been found to interact with the pregnane X receptor. Keywords: natural products , nuclear receptors, pregnane X, xenobiotics. As a result of their work on the pregnane X receptor (PXR, NR1I2), Matic et al. (2007) aptly dubbed it a “promiscuous regulator of detoxification pathways” [1]. Indeed, this short, clear definition of PXR, which belongs to the much larger class of nuclear receptors, is a perfect description. When activated by its ligands, the receptor heterodimerizes with retinoid receptor (RXR), binds to DNA, and induces the expression of genes that encode the synthesis of cytochrome P450 (CYP) monooxygenases. Interestingly, both CYPs and PXR are co-localized in many types of tissue, including the liver, intestine, and kidney [1,2]. It is well-known that drug detoxification begins with a hepatic metabolism carried out by CYPs, which cause chemical modifications to xenobiotics through the use of oxygen and a reduced flavoprotein [3]. This process also affects endogenous mediators such as steroid hormones, bile acids, and several other lipids, including eicosanoids. The most important human CYP is CYP3A4, mainly because of its abundance in human hepatocytes and the variety of its susceptible substrates [4]. For its part, PXR activation is responsible for the increased expression of phase II conjugating enzymes and phase III transporters [5]. While the term pregnane refers to the 17-ethyl-steroid skeleton shared by some of the substrates metabolized, the letter X means that this is an orphan receptor, that is, without a known endogenous ligand. Teleologically speaking, CYPs are cleansing agents because they generally help eliminate potentially toxic substances. However, given that newly generated products can sometimes be highly toxic, activators of PXR expression may actually produce deleterious effects. Such is the case of troglitazone, a tocopherol-like agonist of the family of peroxisome proliferator-activated receptors (PPARs), which was introduced, and then later withdrawn, as an antidiabetic drug [6]. Perhaps one of the most striking features of PXR is the vast range of compounds that can serve as ligands for it. In fact, there is no prerequisite chemical structure or function for a given substance to bind and activate the receptor. Some synthetic hormonal steroids, such as dexamethasone, mifepristone and pregnenolone-16-α-carbonitrile (PCN); or antibiotics, like rifampicin, were promptly described as PXR ligands [7]. 2124 Natural Product Communications Vol. 3 (12) 2008 According to the reviews by Staudinger et al. [8] and by Chang and Waxman [9], several natural products, both molecular entities and herbal products (sic), were reported to interact with PXR, mostly as activators. Some of these compounds are wellknown drugs, such as paclitaxel, the antitumor principle from Taxus brevifolia (Taxaceae); artemisinin, a sesquiterpenoid from Artemisia annua (Asteraceae); and forskolin, a diterpenoid from Coleus forskohlii (Lamiaceae). Others are active ingredients of pharmacologically active extracts, including hyperforin from Hypericum perforatum (Hypericaceae) and both E- and Z-guggulsterones from Commiphora mukul (Burseraceae). Among the very few PXR-antagonists, ecteinascidin-743 (Yondelis®), a complex phenolic tris-isoquinoline from the tunicate Ecteinascidia turbinata, deserves special mention. 2. Natural activators of PXR 2.1. Diterpenoids: Cafestol is a furanoditerpene alcohol (Figure 1) present in Coffea seeds, and therefore also in some drinking preparations, such as the unfiltered coffees of Turkey and Scandinavia. Among the biologically significant activities of this compound, increasing blood cholesterol [10], enhancing glutathione-based detoxification [11], and inhibiting oxidative damage to DNA [12], are several of the best-known. Studies on gene expression related to lipid metabolism, for example, showed that cafestol down-regulates cholesterol 7α-hydroxylase (CYP7A1), thereby repressing bile acid synthesis [13]. For our purposes, it is even more important to note that homozygous deletion in the human CYP7A1 gene causes pronounced hypertriglyceridemia and LDL-hypercholesterolemia [14]. Indeed, Ricketts et al. found that, at 20 μM, cafestol activates PXR in HepG2 cells, with no significant effects on other nuclear receptors such as RXRs, glucocorticoid receptors, or the constitutive androstane receptor (CAR) [15]. The effects of cafestol were not thoroughly extended throughout the body; in fact, no induction of CYP3A11 was detected in mouse livers either 3, 7, or 14 days after administration, whereas a nearly fourfold increase was found in intestinal tissues. However, the expression of the detoxificant enzyme glutathione-S-transferase μ1 (GSTμ1) increased in both the liver and the intestine. Interestingly, in contrast with the standard PXR agonist, PCN, the effect of cafestol on liver GSTμ1 was independent of Máñez Figure 1: Cafestol PXR. As for lipid metabolism, cafestol induced ATPbinding cassette (ABC) transporter type A1 (ABCA1), a protein that is responsible for transporting cholesterol from enterocytes into the circulating blood [15]. This may partially explain why this diterpene markedly raises blood cholesterol levels. 2.2. Phenolic compounds 2.2.1. Phenylpropanoids Coumarins: Many plant coumarins possess an isoprene-derived pyran or furan cycle fused to the benzo-α-pyrone basic structure, leading to the socalled pyranocoumarins or furanocoumarins. Such compounds are quite toxic and have limited pharmacological value, with the exception of 8methoxypsoralen (Figure 2), which is used in combination with UV radiation for the treatment of psoriasis and certain malignant dermatosis. Figure 2: 8-Methoxypsoralen This compound causes different metabolism-related interactions with other drugs, such as nicotine [16] and estrogens [17], and induces CYP1A1-mRNA activation in rat hepatocytes. However, it is also known to markedly inhibit CYP1A1 catalytic activity [18]. The involvement of PXR in the induction not only of CYPs, but also of carboxyl esterase 2 (HCE2) by 8-methoxypsoralen has been assessed with the aid of both overexpression and knockdown of PXR in human hepatoma (Huh7) cells. In cells transfected with the siPXR construct, CYP3A4-, HCE2-mRNA, and PXR levels were reduced, whereas PXR overexpression led to an increase in these indicators [19]. Flavonoids and lignans: In a survey examining the possible activity of several well-known phenolics, Kluth et al. (2007) reported that 25 µM of quercetin (3,5,7,3’,4’-pentahydroxyflavone), one of the most widespread flavonols, activated the expression of Interaction of natural products with pregnane X receptor CYP3A4 in HepG2 cells transfected with the reporter gene construct pGL3-CYP3A4-9. Its effect was higher than that of the standard PXR activator rifampicin [20]. However, quercetin did not increase PXR-mediated gene expression, a finding that is in disagreement with a previous report characterizing the compound as a low-grade PXR activator [21]. A moderate activation of PXR occurred with secoisolariciresinol at 10 µM [21]. This latter compound is a dibenzylbutane lignin present in flaxseeds and other plant products in glycosidic form. 2.2.2. Alkylphenols For the purpose of this review, this group includes several non-phenylpropanoid phenols reputed to be chemopreventive agents. As weakly suggested by this label, these are cancer preventive substances, generally of natural origin, which inhibit tumorigenesis through various mechanisms: inhibition of proinflammatory transcription factors, proliferation of malignant cells, synthesis of antiapoptotic proteins, leukocyte adhesion, angiogenesis, etc [22]. Two of the most widely analyzed alkylphenols, curcumin (diferuloylmethane, from turmeric, Figure 3) and resveratrol (3,5,4’-trihydroxy-transstilbene, from red grapes), activated PXR-mediated gene expression. Figure 3: Curcumin. At 25 µM, this effect was almost three times higher for curcumin than for resveratrol. Both compounds were also found to increase CYP3A4 promoter activity, albeit to a lesser extent than quercetin (see the epigraph “Flavonoids and lignans” [21]). 3. Natural inhibitors of PXR 3.1. Steroids Plant sterols, for example, stigmasterol (Figure 4), campesterol, and sitosterol, are widely considered to be beneficial dietary constituents because they allegedly lower cholesterolemia. However, in certain cases, the effects may be surprisingly contrary. When there is a mutation in the genes regulating the Natural Product Communications Vol. 3 (12) 2008 2125 Figure 4: Stigmasterol synthesis of the ABC-type transporters G5 or G8, high sterolemia can result, causing severe coronariopathy. Furthermore, feeding babies with sterol-rich soy products can lead to cholestasis and hepatic failure [23]. Given that some nuclear receptors are of key importance in the control of bile acid toxicity, Carter et al. investigated the possible interaction with farnesoid X receptor (FXR) and other constructs containing ligand binding domains of another six nuclear receptors. In the context of the present review, it is noteworthy that stigmasterol acetate suppressed the activity of the PXR ligand binding domain in HepG2 cells, with notable specificity [24]. This set of experiments reveals how sterols can be toxic by depriving the liver of detoxification pathways based on nuclear receptors. 3.2. Phenolic compounds Unlike the vast majority of naturally occurring coumarins (benzo-α-pyrones), which are derived from ortho-hydroxycinnamic acid, coumestrol (Figure 5) and its related coumarins present a 4-phenyl substitution. These types of compounds are also known as neoflavones because they can be formally considered as isomers of flavones (2-phenyl-benzo-γ-pyrones). As is the case with many isoflavones (3-phenylbenzo-γ-pyrones), coumestrol possesses estrogenic properties, hence the name. In addition, this agent is a PXR antagonist, as proven by Wang et al. These researchers found that coumestrol reduced the basal reporter activity, as measured by the xenobiotic responsive enhancer module (XREM)-luciferase, in the PXR transiently transfected CV-1 cells by 20% at a dose of 25 µM. While many other nuclear receptors were not sensitive to the compound, ERα and ERβ were highly activated. The activation of human PXR by rifampicin was also inhibited by coumestrol, albeit not by its dimethyl or diacetyl derivatives. These results indicate that the presence of the two free 2126 Natural Product Communications Vol. 3 (12) 2008 Figure 5: Coumestrol phenolic hydroxyls is of the utmost importance. From the results of the dose-response studies, the IC50 value was calculated to be 12 µM. It was also confirmed that coumestrol competes for the ligand-binding domain (Ki = 37 µM) with the PXR agonist SR12813 (4-[2,2-bis(diethoxyphosphoryl)-ethenyl]-2,6-ditertbutylphenol). Gene expression studies for CYP3A4 and CYP2B6 in primary cultures of hepatic cells from two human donors helped determine that 25 µM of coumestrol was able to abolish the induction brought about by SR12813 or rifampicin [25]. 3.3. Sulfur compounds Sulforaphane (Figure 6) is the trivial name of 1-isothiocyanato-4-(methylsulphinyl)-butane, a linear sulfur compound derived from the hydrolysis of glucoraphanin, a glucosinolate found in many cruciferous plants (e.g. the genus Brassica), specially in broccoli sprouts. Figure 6: Sulforaphane It had previously been shown that sulforaphane strongly reduces CYP3A4 mRNA in human hepatocytes, an effect which, in addition to others such as the inhibition of histone deacetylases or the induction of glutathione transferases, may be implicated in the experimental anticancer effect of the drug [26]. Zhou et al. [27] have recently shown how PXR inactivation correlates to the inhibition of CYP3A4 expression. Indeed, at 1 µM, sulforaphane inhibited the reporter activity of the PXR ligands mifepristone and rifampicin; at 25 µM the inhibition of mifepristone was complete. After transfection of HepG2 cells with a GAL4-PXR vector, sulforaphane inhibited GAL4 reporter activity with an IC50 value of 14 µM. Moreover, it was demonstrated that the test compound binds to the ligand binding domain of PXR with a Ki of 16 µM. The authors also studied several related isothiocyanates present in Brassicaceae, including iberin (1- Máñez isothiocyanato-3-(methylsulfinyl)propane), cheirolin (1-isothiocyanato-3-(methylsulfonyl)propane), and erucin (1-isothiocyanato-4-(methylthio)butane). Iberin and cheirolin had roughly the same potency as sulforaphane in repressing the PXR-mediated CYP3A4-luciferase reporter activity in HepG2 cells [27]. 4. Concluding remarks Although the restricted scope of this review prevents us from making too many general assumptions, several interesting points should be emphasized. Vegetal secondary metabolites are xenobiotics for mammalian organisms, which is not surprising since a long list of microbial and plant principles show interaction with PXR; at least as activator ligands. Indeed, the latest studies have added even more compounds to the list of novel PXR inhibitors, which have been dubbed, perhaps inappropriately, “antagonists.” As there is no chemical structure that determines the binding of PXR to date, there is likewise no particular chemical clue which would as yet indicate either the activation or inhibition of such a receptor. As noted above, it is widely thought that PXR not only regulates the detoxification of many structurally unrelated foreign agents, but that it also participates in the metabolism of endogenous steroids. In this context, it is surprising that so few plant steroids and triterpenoids have been studied. In fact, since the discovery of guggulsterones, no new studies have been published in this highly interesting field. For now, the therapeutic value of PXR activators or inhibitors remains obscure. We refer to drugs that owe their application to interactions with PXR, but not to other, some times very important (paclitaxel, rifampicin) drugs that, apart from their main action, have been shown to be notable PXR activators. On the other hand, the role as pharmacologically active ingredients of foods, such as sulforaphane, deserves careful attention. Acknowledgment - The author is indebted with Spanish Ministry of Science for financial support (Project SAF2006-06726) and Laura Gatzkiewicz for her English revision. Interaction of natural products with pregnane X receptor Natural Product Communications Vol. 3 (12) 2008 2127 References [1] Matic M, Mahns A, Tsoli M, Corradin A, Polly P, Robertson GR. (2007) Pregnane X receptor: promiscuous regulator of detoxification pathways. International Journal of Biochemistry and Cellular Biology, 39, 478-483. [2] Willson TM, Kliewer SA. (2002) PXR, CAR and drug metabolism. Nature Reviews Drug Discovery, 1, 259-266. [3] Anzenbacher P, Anzenbacherová E. (2001) Cytochromes P450 and metabolism of xenobiotics. Cellular and Molecular Life Sciences, 58, 737-747. [4] Guengerich FP. (1999) Cytochrome P-450 3A4: regulation and role in drug metabolism. Annual Review of Pharmacology and Toxicology, 39, 1-17. [5] Orans J, Teotico DG, Redinbo MR. (2005) The nuclear xenobiotic receptor pregnane X receptor: recent insights and new challenges. Molecular Endocrinology, 19, 2891-2900. [6] He K, Talaat RE, Pool WF, Reily MD, Reed JE, Bridges AJ, Woolf TF. (2004) Metabolic activation of troglitazone: identification of a reactive metabolite and mechanisms involved. Drug Metabolism and Disposition, 32, 639-646. [7] Kliewer SA, Willson TM. (2002) Regulation of xenobiotic and bile acid metabolism by the nuclear pregnane X receptor. Journal of Lipid Research, 43, 359-364. [8] Staudinger JL, Ding X, Lichti K. (2006) Pregnane X receptor and natural products: beyond drug-drug interactions. Expert Opinion on Drug Metabolism & Toxicology, 2, 847-857. [9] Chang TK, Waxman DJ. (2006) Synthetic drugs and natural products as modulators of constitutive androstane receptor (CAR) and pregnane X receptor (PXR). Drug Metabolism Reviews, 38, 51-73. [10] Weusten-Van der Wouw MP, Katan MB, Viani R, Huggett AC, Liardon R, Liardon R, Lund-Larsen PG, Thelle DS, Ahola I, Aro A, Meyboom S, Beynen AC (1994) Identity of the cholesterol-raising factor from boiled coffee and its effects on liver function enzymes. Journal of Lipid Research, 35, 721-733. [11] Huber WW, Prustomersky S, Delbanco E, Uhl M, Scharf G, Turesky RJ, Thier R, Schulte-Hermann R. (2002) Enhancement of the chemoprotective enzymes glucuronosyl transferase and glutathione transferase in specific organs of the rat by the coffee components kahweol and cafestol. Archives of Toxicology, 76, 209-217. [12] Lee KJ, Jeong HG. (2007) Protective effects of kahweol and cafestol against hydrogen peroxide-induced oxidative stress and DNA damage. Toxicology Letters, 173, 80-87. [13] Post SM, de Roos B, Vermeulen M, Afman L, Jong MC, Dahlmans VE, Havekes LM, Stellaard F, Katan MB, Princen HM. (2000) Cafestol increases serum cholesterol levels in apolipoprotein E*3-Leiden transgenic mice by suppression of bile acid synthesis. Arteriosclerosis, Thrombosis and Vascular Biology, 20, 1551-1556. [14] Pullinger CR, Eng C, Salen G, Shefer S, Batta AK, Erickson SK, Verhagen A, Rivera CR, Mulvihill SJ, Malloy MJ, Kane JP. (2002) Human cholesterol 7α-hydroxylase (CYP7A1) deficiency has a hypercholesterolemic phenotype. Journal of Clinical Investigation, 110, 109-117. [15] Ricketts ML, Boekschoten MV, Kreeft AJ, Hooiveld GJ, Moen CJ, Müller M, Frants RR, Kasanmoentalib S, Post SM, Princen HM, Porter JG, Katan MB, Hofker MH, Moore DD. (2007) The cholesterol-raising factor from coffee beans, cafestol, as an agonist ligand for the farnesoid and pregnane X receptors. Molecular Endocrinology, 21, 1603-1616. [16] Koenigs LL, Trager WF. (1998) Mechanism-based inactivation of P450 2A6 by furanocoumarins. Biochemistry, 37, 10047-10061. [17] Diawara MM, Kulkosky PJ. (2003) Reproductive toxicity of the psoralens. Pediatric Pathology & Molecular Medicine, 22, 247-258. [18] Baumgart A, Schmidt M, Schmitz HJ, Schrenk D. (2005) Natural furocoumarins as inducers and inhibitors of cytochrome P450 1A1 in rat hepatocytes. Biochemical Pharmacology, 69, 657-667. [19] Yang J, Yan B. (2007) Photochemotherapeutic agent 8-methoxypsoralen induces cytochrome P450 3A4 and carboxylesterase HCE2: evidence on an involvement of the pregnane X receptor. Toxicological Sciences, 95, 13-22. [20] Kluth D, Banning A, Paur I, Blomhoff R, Brigelius-Flohé R. (2007) Modulation of pregnane X receptor- and electrophile responsive element-mediated gene expression by dietary polyphenolic compounds. Free Radical Biology & Medicine, 42, 315-325. [21] Jacobs MN, Nolan GT, Hood SR. (2005) Lignans, bacteriocides and organochlorine compounds activate the human pregnane X receptor (PXR). Toxicology & Applied Pharmacology, 209, 123-133. [22] Aggarwal BB, Shishodia S. (2006) Molecular targets of dietary agents for prevention and therapy of cancer. Biochemical Pharmacology, 71, 1397-1421. 2128 Natural Product Communications Vol. 3 (12) 2008 Máñez [23] Berge KE, Tian H, Graf GA, Yu L, Grishin NV, Schultz J, Kwiterovich P, Shan B, Barnes R, Hobbs HH. (2000) Accumulation of dietary cholesterol in sitosterolemia caused by mutations in adjacent ABC transporters. Science, 290, 1771–1775. [24] Carter BA, Taylor OA, Prendergast DR, Zimmerman TL, Von Furstenberg R, Moore DD, Karpen SJ. (2007) Stigmasterol, a soy lipid-derived phytosterol, is an antagonist of the bile acid nuclear receptor FXR. Pediatric Research, 62, 301-306. [25] Wang H, Li H, Moore LB, Johnson MD, Maglich JM, Goodwin B, Ittoop OR, Wisely B, Creech K, Parks DJ, Collins JL, Willson TM, Kalpana GV, Venkatesh M, Xie W, Cho SY, Roboz J, Redinbo M, Moore JT, Mani S. (2008) The phytoestrogen coumestrol is a naturally occurring antagonist of the human pregnane X receptor. Molecular Endocrinology, 22, 838-857. [26] Mahéo K, Morel F, Langouët S, Kramer H, Le Ferrec E, Ketterer B, Guillouzo A. (1997) Inhibition of cytochromes P-450 and induction of glutathione S-transferases by sulforaphane in primary human and rat hepatocytes. Cancer Research, 57, 3649-3652. [27] Zhou C, Poulton EJ, Grün F, Bammler TK, Blumberg B, Thummel KE, Eaton DL. (2007) The dietary isothiocyanate sulforaphane is an antagonist of the human steroid and xenobiotic nuclear receptor. Molecular Pharmacology, 71, 220-229. NPC Natural Product Communications Natural Products as Gastroprotective and Antiulcer Agents: Recent Developments 2008 Vol. 3 No. 12 2129 - 2144 Rosa Tundisa,*, Monica R Loizzoa, Marco Bonesia, Federica Menichinib, FilomenaConfortia, Giancarlo Stattia and Francesco Menichinia a Department of Pharmaceutical Sciences, Faculty of Pharmacy, Nutritional and Health Sciences, University of Calabria, I-87030 Arcavacata di Rende (CS) Italy b Pharmaceutical Sciences Research Division, King’s College London, 150 Stamford Street, London SE1 9NH, UK tundis@unical.it Received: May 27th, 2008; Accepted: October 3rd, 2008 Peptic ulcer, one of the most common gastrointestinal diseases, is a chronic inflammatory disease characterized by ulceration in the regions of the upper gastrointestinal tract where parietal cells are found and where they secrete hydrochloric acid and pepsin. The anatomical sites where ulcer occurs commonly are stomach and duodenum, causing gastric and duodenal ulcer, respectively. Physiopathology of ulcer is due to an imbalance between aggressive factors, such as acid, pepsin, Helicobacter pylori and non-steroidal anti-inflammatory agents, and local mucosal defensive factors, such as mucus bicarbonate, blood flow and prostaglandins. Several drugs are widely used to prevent or treat gastro-duodenal ulcers. These include H2-receptor antagonists, proton pump inhibitors and cytoprotectives. Due to problems associated with recurrence after treatment, there is therefore the need to seek alternative drug sources against ulcers. In recent years, a widespread search has been launched to identify new gastroprotective drugs from natural sources. The aim of the present review is to highlight the recent advances in current knowledge on natural products as gastroprotective and antiulcer agents and consider the future perspectives for the use of these compounds. Keywords: gastroprotective agents, plant extracts, terpenes, flavonoids, xanthones. Peptic ulcer disease is a problem of the gastrointestinal tract characterized by mucosal damage secondary to pepsin and gastric acid secretion. It usually occurs in the stomach and proximal duodenum; less commonly, it occurs in the lower esophagus, the distal duodenum or the jejunum, as in hypersecretory states, hiatal hernias or ectopic gastric mucosa. Helicobacter pylori infection and the use of non-steroidal anti-inflammatory drugs (NSAIDs) are the predominant causes of peptic ulcer disease in the United States [1]. H. pylori infection leads to gastroduodenal inflammation, peptic ulceration, gastric lymphoma, and gastric cancer, which has been proven with animal studies and human epidemiological report. H. pylori may induce inflammatory-associated gene expression in gastric epithelial cells, including activation of nuclear factor kappa B (NF-κB), enhance expression of cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS), and production of interleukin-8 (IL-8). H. pylori bacteria adhere to the gastric mucosa; the presence of another inflammatory protein and a functional cytotoxinassociated gene island in the bacterial chromosome increases virulence and probably ulcerogenic potential [2]. NSAIDs can cause damage to the gastro-duodenal mucosa via several mechanisms, including their topical irritant effect on the epithelium, impairment of the mucosal barrier function, suppression of gastric prostaglandin synthesis, reduction of gastric mucosal blood flow, and interference with the repair of superficial injury. The presence of acid in the lumen of the stomach also contributes to the pathogenesis of NSAIDs-induced ulcers and bleeding by impairing the restitution process, interfering with hemostasis and inactivating several growth factors that are important in mucosal 2130 Natural Product Communications Vol. 3 (12) 2086 defense and repair [3]. A variety of other infections and co-morbidities are associated with a greater risk of peptic ulcer disease, such as Crohn’s disease, hepatic cirrhosis, cytomegalovirus, tuberculosis, chronic renal failure, sarcoidosis and myeloproliferative disorder. Most of the available gastroprotective drugs act on the offensive factors neutralizing acid secretion, like antacids, H2 receptor blockers, like ranitidine, anticholinergics, like pirenzepin, proton pump inhibitors, like omeprazole, and lansoprazole, which interfere with acid secretion. However, the use of these antisecretory drugs may be associated with adverse events and ulcer relapse [4]. Thus, there is a need for more effective, less toxic and cost-effective anti-ulcer agents. Herbal medicines have been used since the dawn of civilization to maintain health and to treat diseases. The World Health Organization estimates that about three quarters of the world’s population currently use herbs and other forms of traditional medicines to treat their diseases because the use of these compounds are considered as safe [5,6]. In recent years, a widespread search has been launched to identify new anti-ulcer drugs from natural sources. In traditional medicine, for example, several plants have been used to treat gastrointestinal disorders, including gastric ulcers [7-9]. The potential use of plants has been successfully demonstrated in the field of gastroprotection in a recent article that reviewed the studies on extracts and pure compounds as gastroprotective agents reported in the literature up to 2005 [10]. The purpose of the present review article is to highlight the more recent data in current knowledge on natural products as gastroprotective and antiulcer agents. The mechanism of action and the structureactivity relationships are also discussed where it is possible. 1. Extracts Several studies on the gastroprotective effects of plant extracts have been recently undertaken. In a recent study, de Andrade et al. [11] evaluated the effects of Maytenus robusta extract, a plant used in folk medicine for the treatment of stomach ulcers, using the NSAIDs-induced ulcer, ethanol-induced ulcer and stress-induced ulcer protocols. Tundis et al. In the ethanol-induced ulcer model, it was observed that the treatment with M. robusta extract (50, 250 and 500 mg/kg) and positive control omeprazole (30 mg/kg) significantly reduced the lesion index, the total lesion area and the percentage of lesion, in comparison with the negative control group. The percentages of inhibition of ulcers were 75.1, 85.0, 86.6 and 75.5 for the treated groups with 50, 250 and 500 mg/kg of M. robusta and omeprazole, respectively. Significant inhibition was also observed in the lesion index in the indomethacin-induced ulcer model, the decrease being 62.5, 62.5, 63.6 and 96.2 for groups treated with 50, 250 and 500 mg/kg of Maytenus robusta and positive control (cimetidine), respectively. Similar results were observed in the stress-induced ulcer model, where the inhibition of ulcer lesions was 71.3, 72.7, 76.5 and 92.3 for the groups treated with 50, 250 and 500 mg/kg of plant extract. Regarding the model of gastric secretion, a reduction in the volume of gastric juice volume and total acidity was observed, as well as an increase in gastric pH. At 200 mg/kg body weight (b.w.) the aqueous extract of Decalepis hamiltonii protected swim stressinduced ulcer lesions by 77%, similar to that of ranitidine (79%), a known antiulcer drug, at 30 mg/kg b.w. [12]. Reactive oxygen species (ROS) have been implicated in the pathogenesis of a wide variety of clinical disorders and gastric damage. Preventive antioxidant enzymes such as superoxide dismutase (SOD) and catalase (CAT) are the first line of defence against ROS. Administration of D. hamiltonii extract resulted in a significant increase in the SOD, catalase and reduced glutathione (GSH) levels, similar to those of control animals, suggesting the efficacy of D. hamiltonii extract in preventing free radical-induced damage during ulceration. The extract also normalized the approximately3.1 and 2.4 folds of increased H+-K+-ATPase and gastric mucin, respectively, in ulcerous animals, to levels similar to those found in healthy controls. The gastroprotective effects of an aqueous suspension of the ethanolic extract of leaves and flowers of Guazuma ulmifolia was assessed in a model of acute gastric ulcer induced by diclofenac, using the proton pump inhibitor omeprazole as a protection reference [13]. Pretreatment with G. ulmifolia decreased the ulcerated area by diclofenac in a dose-dependent way. Myeloperoxidase activity, as a marker of neutrophil infiltration, was slightly reduced in vivo, whereas in vitro anti-inflammatory Gastroprotective natural products Natural Product Communications Vol. 3 (12) 2008 2131 activity was clearly inhibited in a dose-dependent manner. The lowest doses of the extract significantly decreased the levels of lipoperoxides, and superoxide dismuthase activity increased to a similar extent as with omeprazole. Examination of glutathione metabolism reflected a significant rise in glutathione peroxidase (GPx) activity at the highest dose of G. ulmifolia. These results showed that the aerial parts of G. ulmifolia had demonstrated protection of the gastric mucosa against the injurious effect of NSAIDs, mainly by anti-inflammatory and radical scavenging mechanisms. cells. The results indicate that the gastroprotective effect of T. arjuna extract is probably related to its ability to maintain the membrane integrity by its antilipid peroxidative activity that protects the gastric mucosa against oxidative damage and its ability to strengthen the mucosal barrier, the first line of defense against exogenous and endogenous ulcerogenic agents. A crude hydroalcoholic extract of Polygala paniculata administered orally was able to protect the gastric mucosa against lesions induced by ethanol 70% [14]. In this study the extract was given by two routes (oral and intraperitoneal) to evaluate whether the observed effect was related to an adherent property of the extract on the gastric mucosa by forming a protective barrier against the aggressive effects of ethanol. The results showed that, when given by the intraperitoneal route, P. paniculata extract exhibited an important cytoprotective effect similar to the one seen when the extract was given orally. This suggested that the pharmacological mechanism did not have any relationship with an adherent property of the extract. In addition, the extract partially protected the mucosa against indomethacin-induced lesions. The extract did not change the volume and acidity of gastric secretion and exerted an antioxidant activity. The gastroprotective effects of P. paniculata extract may have involved prostaglandins and be related to cytoprotective factors, such as antioxidant activity and maintenance of mucus production. The methanol extract of the bark of Terminalia arjuna showed marked antiulcer and ulcer healing activity against 80% ethanol, diclofenac sodium and dexamethasone induced ulcer models dosedependently [15]. Pre-, post and co-administration of extract showed 100% protection to the gastric mucosa against ethanol, diclofenac and dexamethasone induced ulcers. T. arjuna increased the levels of GSH in gastric mucosa, which were reduced upon ulcer induction with dexamethasone. These results suggest that GSH depletion has a role to play in the ulcerogenesis induced by dexamethasone. The restoration of GSH levels by the extract provides evidence for the involvement of GSH in the antiulcer activity of T. arjuna. T. arjuna also increased significantly the glycoprotein content of the mucosal Some spices, namely black pepper, ginger, and turmeric have been shown to possess significant gastroprotective activities [16-18]. Recently, Al Mofleh et al. [19] provided substantial evidence for anti-ulcer and anti-secretory effects of an aqueous suspension of anise (Pimpinella anisum). Anise suspension significantly inhibited the ulcerative lesions in all animals treated with necrotizing agents. Chemical studies demonstrated that P. anisum and its major constituents, anethol, eugenol, anisaldehyde, methylchanicol, other terpenes and coumarins, were free radicals or active oxygen scavengers. In addition, the ability of anise suspension to protect gastric mucosa against lesions induced by chemical irritants is likely by maintaining the structural integrity of the gastric epithelium and a balance between aggressive factors and inherent protective mechanisms. Previously, the same research group evaluated the anti-ulcerogenic property of an aqueous suspension of Mentha piperita in different ulcer models in vivo [20]. The suspension at 250 and 500 mg/kg b.w., orally (i.p. in Shay rat model), had a significant effect in pyloric ligation induced basal gastric secretion, in indomethacin and noxious chemical (80% ethanol, 0.2 M NaOH and 25% NaCl) induced gastric ulceration. The aqueous suspension showed significant protection in all models used. These findings were supported by histopathological assessment of gastric tissue and by the determination of non-protein sulfhydryl (NP-SH) contents of the stomach, as these parameters showed protection of various indices and replenishment of the depleted NP-SH level by the suspension treatment, respectively. The ulcer protective effect of M. piperita may possibly be due to its anti-secretory effect, along with antioxidative and cytoprotective properties through a prostaglandins mediated mechanism. The effect of Carum carvi pretreatment on gastric mucosal injuries caused by NaCl, NaOH, ethanol and pylorous ligation accumulated gastric acid secretions was investigated in rats [21]. Pretreatment at oral doses of 250 and 500 mg/kg b.w. was found to 2132 Natural Product Communications Vol. 3 (12) 2086 provide a dose-dependent protection against the ulcerogenic effects of different necrotizing agents, ethanol-induced histopathological lesions, depletion of stomach wall mucus and NP-SH groups and pylorous ligated accumulation of gastric acid secretions. The protective effect of C. carvi against ethanol-induced damage of the gastric tissue appears to be related to the free-radical scavenging property of its constituents. The exact mechanism of action of the gastroprotective activity is not known. However, it might be due to flavonoid related suppression of cytochrome P450 1A1 (CYP1A1), which is known to convert xenobiotics and endogenous compounds to toxic metabolites. Another spice, Coriandrum sativum, was evaluated for its gastroprotective activity [22]. Pretreatment at oral doses of 250 and 500 mg/kg b.w. was found to provide a dose-dependent protection against the ulcerogenic effects of different necrotizing agents, ethanol-induced histopathological lesions, pylorus ligated accumulation of gastric acid secretions and ethanol related decrease of NP-SH. Results obtained from the study of gastric mucus and indomethacininduced ulcers demonstrated that the gastroprotective activity of C. sativum might not be mediated by gastric mucus and/or endogenous stimulation of prostaglandins, but might be related to the freeradical scavenging property of different antioxidant constituents (coumarins, catechins, terpenes and polyphenolic compounds) present in C. sativum. The inhibition of ulcers might be due to the formation of a protective layer of either one or more than one of these compounds by hydrophobic interactions. An aqueous suspension of Crocus sativus was evaluated in rats for its gastric antiulcer activity induced by pylorus ligation, indomethacin and various necrotizing agents, including 80% ethanol, 0.2 M NaOH and 25% NaCl [23]. Gastric wall mucus and NP-SH contents were also estimated in rats. Histopathological assessment of the stomach was carried out. The C. sativus aqueous suspension at doses of 250 and 500 mg/kg exhibited decreases in basal gastric secretion and ulcer index in Shay rats and indomethacin treated groups. Gastric wall mucus elevation was observed, but no significant histopathological changes were noted. C. sativus exhibited significant antisecretory and antiulcer activities without causing any deleterious effects on acute and chronic toxicity in rodents. Tundis et al. In the ethanol-induced stress gastric ulcer test in rats, it was shown that the Carlina acanthifolia essential oil, traditionally used in the treatment of various disorders, including stomach diseases, produced significant dose-dependent gastroprotective activity [24]. This was particularly noticeable when the essential oil was given as a “pure oil” in a dose of 1.0 mL/kg. The free radical scavenging activity of the essential oil tested might be considered as one of the possible mechanisms of the gastroprotective effect observed [25]. The effect of San-Huang-Xie-Xin-Tang (a traditional oriental medicinal formula containing Coptis chinesis, Scutellaria baicalensis and Rheum officinale) and its main component baicalin was recently evaluated on H. pylori-infected human gastric epithelial AGS cells [26]. It is widely accepted that most peptic ulcers are associated with H. pylori infection and eradication of the organism leads to enhanced ulcer healing and reduces the chance of ulcer recurrence. NF-κB activation plays an important role in H. pylori-induced inflammation and apoptosis in gastric epithelial cells [27]. Treatment with San-Huang-Xie-Xin-Tang and baicalin significantly inhibited IκBα degradation and NF-κB activation in H. pylori-infected AGS cells. Previously, H. pylori-induced inflammation has been shown to be associated with COX-2 expression in experimental animals and human patients [28]. Furthermore, in early gastric cancer and intestinal metaplasia the expression of COX-2 in patients infected by H. pylori is increased [29]. Thus, chronic expression of COX-2 may play an important role in H. pylori-associated gastric carcinogenesis, in addition to propagation of gastric inflammation [30]. San-Huang-Xie-Xin-Tang and baicalin decreased H. pylori-induced COX-2 expression in human gastric epithelial cells. Thereby, they might inhibit COX-2 associated gastric inflammation. Recent studies also demonstrated that H. pylori acts through TLR2/TLR9 to activate both the PI-PLCγ/PKCα/c-Src/IKKα/β and NIK/IKKα/β pathways, resulting in the phosphorylation and degradation of IκBα, which in turn leads to the stimulation of NF-κB and COX-2 gene expression [31]. Thus, it was suggested that San-Huang-XieXin-Tang and baicalin suppression of COX-2 expression might be mediated via inhibition of degradation of IκBα. Gastroprotective natural products Natural Product Communications Vol. 3 (12) 2008 2133 IL-8 secreted by gastric epithelial cells is likely to be an important host mediator inducing neutrophil migration to the site of infection and, therefore, may be important in the regulation of inflammatory and immune processes in response to H. pylori [32]. SanHuang-Xie-Xin-Tang and its major component also inhibit H. pylori-induced IL-8 production and, therefore, increase the benefit on gastric mucosal protection. 200 mg/kg) against aspirin-induced gastric lesions could possibly be due to its 5-lipoxygenase inhibitory effect. Ethanol-induced depletion of gastric wall mucus has been significantly prevented by A. latifolia extract. Pylorus ligation-induced ulcers are due to auto-digestion of the gastric mucosa and breakdown of the gastric mucosal barrier [36]. A. latifolia extract also protected against the cold-resistant stressinduced ulcers and pylorus ligation at 200 mg/kg. Stress-induced ulcers are probably mediated by histamine release with enhancement in acid secretion and a reduction in mucous production. Increase in gastric motility, vagal overactivity, mast cell degranulation decreased gastric mucosal blood flow, and decreased prostaglandin synthesis is involved in genesis of stress-induced ulcers [37-39]. Accordingly, the protective action of A. latifolia extract against stress-induced ulceration could be due to its histamine antagonistic, anticholinergic and antisecretory effects. Alchornea glandulosa (Euphorbiaceae) is a plant used in folk medicine as an antiulcer agent. Rats pretreated with a methanolic extract of the leaves showed a significant, dose-dependent reduction of gastric ulcers induced by absolute ethanol. Pretreatment of mice with A. glandulosa extract (500, 1000 mg/kg, p.o.) showed significant, dosedependent decreases in the severity of lesions caused by HCl/ethanol and by NSAID-induced gastric lesions. Pretreatment with the extract also induced antisecretory action via local and systemic routes and a significant decrease in the total gastric acid content. The gastroprotective effects of A. glandulosa involved the participation of nitric oxide (NO) and increased levels of endogenous sulfhydryl compounds, which are defensive mechanisms of the gastrointestinal mucosa against aggressive factors. The results showed that single oral administrations of A. glandulosa (250 mg/kg/once daily) potently stimulates gastric epithelial cell proliferation that contributes to the accelerated healing of gastric ulcers induced by acetic acid. In addition, no sub-acute toxicity (body weight gain, vital organs and serum biochemical parameters) was observed during treatment with the extract. Phytochemical investigation led to the isolation of gallic acid, methyl gallate, pterogynidine and different flavonoids. These compounds may contribute to the observed antiulcerogenic effects of A. glandulosa [33]. The gastroprotective potential of the 50% aqueous alcoholic extract of Anogeissus latifolia (100 and 200 mg/kg b.w.) was studied on aspirin, cold-resistant stress, pylorus ligated and ethanol-induced ulcers. The status of the antioxidant enzymes SOD and CAT, along with lipid peroxidation (LPO), was also studied in cold-resistant stress-induced ulcers [34]. Synthetic NSAIDs, like aspirin, cause mucosal damage by interfering with prostaglandin synthesis, increasing acid secretion and back diffusion of H+ ions, resulting in overproduction of leukotrienes and other products of the 5-lipoxygenase pathway [35]. Hence, the protective action of A. latifolia extract (65.6% at Stachytarpheta cayennensis is an herbaceous plant popularly known as gervão-roxo, gervão-do-campo or vassourinha-de-botão in Brazil and used in traditional medicine for the treatment of gastritis and ulcers [40]. Two hydroalcoholic extracts obtained respectively with ethanol/water 70:30 and ethanol/water 96:4 were prepared and tested. The oral pretreatment with the second ethanolic extract significantly inhibited the generation of gastric lesions induced by diclofenac, whereas the first extract induced a slight, but not statistically significant inhibition of mucosa damage. Mouriri pusa is another medicinal plant commonly used in Brazil against gastric ulcer. The methanol and dichloromethane extracts obtained by sequential extraction from the leaves of M. pusa were evaluated for their ability to protect the gastric mucosa against injuries caused by necrotizing agents (0.3M HCl/60% EtOH, absolute ethanol, NSAIDs, stress and pylorus ligature) in mice and rats [41]. The best results were obtained after pretreatment with methanol extract, whereas the dichloromethane extract did not show the same significant antiulcerogenic activity. The mechanism involving the antiulcerogenic action of the methanol extract seemed to be related to NO generation and also suggested the effective participation of endogenous sulfhydryl groups in the gastroprotective action. Phytochemical investigation of the methanol extract of M. pusa yielded tannins and flavonoids. The presence of these phenolic compounds probably would explain the 2134 Natural Product Communications Vol. 3 (12) 2086 antiulcerogenic effect of the polar extract of M. pusa leaves. Recently, Berenguer et al. [42] evaluated the gastroprotective effect of Rhizophora mangle in a model of diclofenac-induced ulcers in rats and studied the mechanisms involved, using the proton pump inhibitor omeprazole for comparison. The major active principles are polyphenols [43]. These compounds have shown cytoprotective properties [44] and have been associated with antiulcerogenic activity in other plants [45,46]. The topical action of the aqueous extract of R. mangle in accelerating wound healing has been previously explained by several mechanisms, such as coating the wound, forming complexes with proteins of the microorganism cell wall, chelating free radicals and reactive oxygen species, stimulating the contraction of the wound and increasing the formation of new capillaries and fibroblasts [47]. Berenguer et al. [42] found a thick coating of R. mangle extract macroscopically adherent to the gastric mucosa, which suggests that in addition to antioxidant mechanisms, the formation of a physical barrier with similar properties as observed in topical wounds may contribute to the gastroprotective action of the drug. A methanolic extract, the essential oil, light petroleum soluble and insoluble fractions of the methanolic extract of Elettaria cardamomum were studied in rats at doses of 100-500, 12.5-50, 12.5-150 and 450 mg/kg, respectively, for their ability to inhibit the gastric lesions induced by aspirin, ethanol and pylorous ligature [48]. In addition, effects on wall mucus and gastric acid output were recorded. All fractions significantly inhibited gastric lesions induced by ethanol and aspirin, but not those induced by pylorus ligation. The methanolic extract proved to be active, reducing lesions by about 70% in the ethanol-induced ulcer model at 500 mg/kg. The light petroleum soluble fraction reduced the lesions by 50% at 50 and 100 mg/kg, with similar effect to that of the insoluble fraction of the methanol extract at 450 mg/kg. In the aspirin-induced gastric ulcer, the best gastroprotective effect was found in the light petroleum soluble fraction, which inhibited lesions by nearly 100% at 12.5 mg/kg. Oral administration of Kaempferia parviflora ethanolic extract (30-120 mg/kg) inhibited gastric ulcer formation induced by indomethacin, HCl/EtOH and water immersion restraint stress [49]. It was Tundis et al. found that pretreatment with K. parviflora at doses of 60 and 120 mg/kg significantly increased the amount of gastric mucus content in HCl/EtOH-ulcerated rats. The finding that K. parviflora failed to increase the gastric pH and decrease the gastric volume and acidity in pylorus-ligated rats suggests that the antisecretory action is unlikely to be ascribed to the anti-gastric ulcer effect of the K. parviflora. The gastric wall mucus is thought to play an important role as a defensive factor against gastrointestinal damage. The gastric wall mucus was used as an indicator for gastric mucus secretion. The finding that pretreatment with K. parviflora at doses of 60 and 120 mg/kg significantly increased gastric mucus content in HCl/EtOH ulcerated rats suggests that the gastroprotective effect of K. parviflora is mediated only partly by preservation of gastric mucus secretion. Sannomiya et al. [50] evaluated the potential antiulcerogenic effect of three different extracts obtained from the leaves of Byrsonima crassa, namely hydromethanolic (80% MeOH), methanolic and chloroform extracts. The oral administration of all the extracts reduced the formation of lesions associated with HCl/ethanol administration in mice. The 80% MeOH extract significantly reduced the incidence of gastric lesions by 74, 78 and 92% at doses of 250, 500 and 1000 mg/kg, respectively. The methanolic extract reduced the ulceration only at doses of 500 and 1000 mg/kg. Phytochemical investigation of B. crassa revealed the presence of phenolic compounds that may probably explain the antiulcerogenic effect of the extracts of B. crassa. In the HCl/EtOH-induced gastric ulcer model, an hydroalcoholic extract obtained from Pradosia huberi barks demonstrated significant inhibition of the ulcerative lesion index by 73% (500 mg/kg) and 88% (1000 mg/kg), respectively [51]. The gastric damage induced by absolute ethanol in rats was effectively reduced by 84, 88 and 81% (250, 500 and 1000 mg/kg). In the NSAID-induced lesion model, P. huberi extract also showed an antiulcerogenic effect with decrease in gastric lesions. P. huberi administered either orally or intraduodenally was able to change gastric juice parameters as well as those treated with cimetidine. The treatment with P. huberi extract significantly increased gastric volume, the pH values and promoted reduced acid output. By comparison of the effects produced by the intraduodenal and oral routes, it was observed that P. huberi was better for local activity in gastric Gastroprotective natural products Natural Product Communications Vol. 3 (12) 2008 2135 mucosa than in systemic action. The hydroalcoholic extract of P. huberi was also shown to be an inhibitor of intestinal motility. The mechanism of action of the.extract did not seem to be related to the NOinhibitor, but showed the participation of endogenous sulphydryl groups in the gastroprotective action. treated groups of animals as compared with the control group. Histopathological examination of the stomach of the ulcerated animals showed severe erosion of the gastric mucosa, sub-mucosal edema and neutrophil infiltration. All of these symptoms were found to be normal in the treated groups. In general, the results of this investigation revealed the gastroprotective activity of the extract through an antioxidant mechanism. In order to establish the pharmacological basis for their ethnomedicinal use in gastric disorders, studies were made of the effects of ethanol extracts and fractions from root tubers of Cynanchum auriculatum, C. bungei and Cynoctonum wilfordii on ethanol- and indomethacin-induced gastric lesions, and histamine-induced gastric acid secretion in rats [52]. Oral administration of the ethanol extract and chloroform fraction of C. wilfordii at doses of 150 and 68 mg/kg, respectively, significantly inhibited the development of ethanol- and indomethacininduced gastric lesions and also caused a significant decrease of gastric acid secretion after histamineinduced gastric lesion. Oral administration of ethanol extract and the chloroform fraction of C. auriculatum at doses of 300 and 69 mg/kg, respectively, significantly inhibited ethanol- and indomethacininduced gastric lesions. Cissus quadrangularis is well known for the treatment of gastric disorders owing to it being a rich source of carotenoids, triterpenoids and ascorbic acid. Jainu et al. [53] evaluated an ethanol extract of C. quadrangularis against the gastric toxicity induced by aspirin in rats with an optimum protective dose of 500 mg/kg of extract in the aspirin model. In addition, administration of aspirin increases lipid peroxidation status, xanthine oxidase (XO), and myeloperoxidase, and decreases selenium-GPx activities in the gastric mucosa, resulting in mucosal damage at both cellular and subcellular level. Pretreatment with C. quadrangularis ameliorated the observed effects significantly in the gastric mucosa of ulcerated rats. These findings suggest that the gastroprotective activity of C. quadrangularis could be mediated possibly through its antioxidant effect, as well as by the attenuation of the oxidative mechanism and neutrophil infiltration. Administration of a 70% methanolic extract of Punica granatum fruit rind showed inhibition in aspirin- and ethanol-induced gastric ulceration [54]. In treated groups of animals, the SOD, CAT, GSH and GPx levels were increased and found more or less equal to the normal values. The tissue lipid peroxidation level was found to be decreased in Al-Qarawi et al. [55] evaluated, in a rat model of ethanol-induced gastric ulceration, the beneficial effects on gastric ulcers of a plant used in folk medicine, Phoenix dactylifera. Aqueous and ethanolic undialyzed and dialyzed extracts from date fruits and pits were given orally to rats at a dose of 4 mL/kg for 14 consecutive days. On the last day of treatment, rats were fasted for 24 h and were then given 80% ethanol (1 mL/rat) by gastric intubation to induce gastric ulcer. Rats were killed after 1 h of ethanol exposure and the incidence and severity of the ulceration were estimated, as well as the concentrations of gastrin in plasma, and histamine and mucus in the gastric mucosa. As a positive control, a single group of rats that were fasted for 24 h was administered orally with lansoprazole and was given 80% ethanol, as above, 8 h thereafter. The results indicated that the aqueous and ethanolic extracts of the date fruit and, to a lesser extent, date pits, were effective in ameliorating the severity of gastric ulceration and mitigating the ethanol-induced increase in histamine and gastrin concentrations, and the decrease in mucin gastric levels. The ethanolic undialyzed extract was more effective than the other extracts used. It is postulated that the basis of the gastroprotective action of P. dactylifera extracts may be multi-factorial, but may include an antioxidant action. The tissue lipid peroxidation level was found to be decreased in the treated groups of animals as compared with the control group. Histopathological examination of the stomach of the ulcerated animals showed severe erosion of the gastric mucosa, submucosal edema and neutrophil infiltration. Portulaca oleracea, commonly used in Iranian folk medicine, has been demonstrated to protect mice from gastric aggressive factors and its administration reduced total gastric acidity and increased pH of gastric juice [56]. On induction of gastric ulceration by using HCl, pretreatment with the aqueous and ethanolic extracts showed a dose-dependent reduction 2136 Natural Product Communications Vol. 3 (12) 2086 Tundis et al. in the severity of ulcers. The dose of 0.8 g/kg of the aqueous extract and 1.4 g/kg of the ethanolic extract had similar activity to sucralfate (0.1 g/kg). In lesions induced by ethanol, the dose of 0.56 g/kg, and 0.8 g/kg of the aqueous extract, and 0.8 and 1.4 g/kg of the ethanolic extract showed significant inhibition of lesions. The oral and intraperitoneal doses of both extracts inhibited the total gastric acidity in the pylorus-ligated mice in a dose-dependent manner. The highest dose of extracts had antisecretory activity, which was comparable to cimetidine. Lavandula hybrida Reverchon “Grosso” exerted gastroprotective effects [57]. Interestingly, the principal constituents of the oil, linalool and linalyl acetate, were demonstrated to contribute to the gastroprotective effect of lavender oil which, orally administered, caused a dramatic reduction in ethanolinduced gastric injury to rats. The lack of a protective effect against gastric mucosal damage caused by indomethacin led to the hypothesis that gastroprotection afforded by L. hybrida oil cannot be attributed to interference with the arachidonic acid metabolic cascade. 1 2 3 4 5 6 7 ROC Of the sesquiterpenes 1-8, assessed at a single oral dose of 50 mg/kg, the best gastroprotective effect was observed for derivative 8, obtained as a diasteromeric mixture by reduction of the 4,5-double bond of cyperenoic acid (3). Compound 8 reduced the lesion index by 86%, being the most active of the sesquiterpenes evaluated in this work and more active than lansoprazole at 20 mg/kg. The products 1 and 3-8 did not show significant differences in gastroprotective activity. Cyperenol (1) and cyperenoic acid methyl ester (4), however, were more cytotoxic with IC50 values of 44 and 75, and 48 and 75 mM against AGS cells and fibroblasts, respectively. The best gastroprotective effect with a H ROC H OH 13 H R = OH HN CH3 14 R = 9 R = OH 10 R = OCH3 11 R = HN 12 R = 8 R = CH2OH R = CH2OCOCH3 R = COOH R = COOCH3 R = CONHCH2CH3 R = CONHCH2CH2CH2CH3 R = CONHPhOCH3 H CH3 HN H OCH3 ROC H 15 R = OH HN 16 R = CH3 CH2OAc CH2OAc 2. Pure compounds 2.1. Terpenes: Several plant terpenoids, including sesquiterpenes, diterpenes and triterpenes, have been shown to protect the gastric mucosa against the damage caused by different ulcerogens [58]. Recently the gastroprotective effect of the sesquiterpene cyperenoic acid and seven semi-synthetic derivatives was assessed in the HCl/ethanol-induced gastric ulcer model in mice [59]. At doses of 25, 50 and 100 mg/kg, cyperenoic acid (3) showed a dose-dependent gastroprotective effect, reducing the ulcers by 45 and 75% at 50 and 100 mg/kg, respectively, compared with the untreated controls. HOOC R HOOC H HOOC 17 H 18 CH2OAc CH2OAc ROC H R= HN D A C ROC R= B 20 21 22 25 27 28 A=B=C=D=H A =B = D = H, C = OCH3 A = C = OCH3, B = D = H A = B = D = H, C = I A = Br, B = C = D = H A = C = D = H, B = Br HN H D A C B 19 23 24 26 29 30 A=B=C=D=H A =B = D = H, C = OCH3 A = C = OCH3, B = D = H A = B = D = H, C = I A = Br, B = C = D = H A = C = D = H, B = Br lower cytotoxicity was found for compound 8, cyperenoic acid (3) and the p-anisidyl derivative 7. The main sesquiterpene of Fabiana imbricata, 11hydroxy-4-amorphen-15-oic acid (9), at doses of 25, 50 and 100 mg/kg showed a dose-dependent gastroprotective effect in HCl/EtOH-induced gastric lesions in mice, reducing the lesions by 68% at 100 mg/kg [60]. Seven derivatives of this terpene were prepared and their gastroprotective effects were assessed in HCl/EtOH-induced gastric lesions in mice. Compounds 9, 10 and 12 reduced the lesion index by 60-65%, while the mixture of compounds 13 and 15, as well as 14 and 16 presented values of 71% and 51%, respectively. The most active compound proved to be the amide derivative 11, Gastroprotective natural products which reduced the lesion index by 80%. At 20 mg/kg, lansoprazole reduced the lesion index by 70%. Compounds 13 and 15, lacking the alcohol function at C-11, had a better gastroprotective activity than 9. In the case of compounds 14 and 16, where the alcohol function at C-11 is absent and the acid group is substituted by an amide function, the gastroprotective activity was reduced when compared with compound 11. The alcohol function at C-11 is required for a better gastroprotective effect when there is a substitution of the acid for an amide. At doses of up to 1000 mg/kg, oral administration of 9 did not show any observable symptoms of toxicity or mortality in mice. Therefore, the intraperitoneal LD50 for this compound in mice is higher than 1000 mg/kg and it can be regarded as ‘not harmful’. The cytotoxicity study revealed that compound 9, as well as the mixtures of 13 and 15 and compounds 14 and 16 presented low toxicity towards AGS cells and fibroblasts. In the compound series 9-12, when the acid function at C-15 is substituted forming an amide (11 and 12), the cytotoxicity increased significantly. A comparison of the acids 9, 13 and 15 indicated a comparable low cytotoxicity, thus suggesting that the presence of the alcohol function at C-11 did not contribute to this effect. When the corresponding amides were prepared (i.e. 11-12 and 14-16), the presence of the hydroxy group at C-11 determined the cytotoxicity of the products. The labdane diterpenes 15-acetoxyimbricatolic acid (17) and 15-acetoxylabd-8(9)-en-19-oic acid (18) isolated from Araucaria araucana exhibited significant gastroprotective activity at 50 and 100 mg/kg in mice, respectively. From these compounds, some aromatic amides were prepared and assessed for their gastroprotective effect in the HCl/EtOH-induced gastric lesion model in mice [61]. The analysis of the gastroprotective activity of the benzylamides belonging to the series 8(9)- and 8(17)-ene was undertaken at doses of 12.5, 25 and 50 mg/kg in the HCI/EtOH-induced gastric lesion model in mice. A significant gastroprotective effect was observed for 15-acetoxylabd-8(9)-en-19-oic acid benzylamide (19) starting at 12.5 mg/kg, reducing the gastric lesions by 50%, while 15-acetoxylabd-8(17)-en-19-oic acid benzylamide (20) reduced lesions by 66% at 25 mg/kg. At 25 mg/kg, the highest gastroprotective effect was observed for the benzyl- and 3-bromo phenylamides from 17, as well as for the benzyl- and Natural Product Communications Vol. 3 (12) 2008 2137 CH2OAc CH2OAc HOOC H HOOC 17 H 18 CH2OAc CH2OAc HN ROC H 20 21 22 25 27 28 R= HN D ROC A A=B=C=D=H A =B = D = H, C = OCH3 A = C = OCH3, B = D = H A = B = D = H, C = I A = Br, B = C = D = H A = C = D = H, B = Br H C B R= D A C B 19 23 24 26 29 30 A=B=C=D=H A =B = D = H, C = OCH3 A = C = OCH3, B = D = H A = B = D = H, C = I A = Br, B = C = D = H A = C = D = H, B = Br p-toluidylamides from 18, these being as active as lansoprazole at 20 mg/kg. The presence of a 4'-methoxy or a 2',4'-dimethoxy functionality did not result in significant differences in the gastroprotective effects of 21 and 22, but a strong effect was observed for 23, while the activity of 24 was lower. The effect of a halogen in the aromatic ring on the gastroprotective activity can be assessed by comparing 25 and 26, which bear iodine. While the gastroprotective activity of 27 and 28 was strong and comparable to that of 20, there was a substantial decrease in the gastroprotective effect of 29 and 30 compared with 19. The results suggest a relevant role of the exomethylene function in the gastroprotective effect of the brominated derivatives, with higher activity for 28. The effect, however, is not statistically different from that of 27. The structural modifications undertaken led to labdane derivatives with an increased gastroprotective effect compared with the parent compounds. The gastroprotective effect of the diterpenes jatropholone A (31), jatropholone B (32) and sixteen semisynthetic derivatives was assessed in the HCl/ethanol-induced gastric lesion model in mice and the cytotoxicity was determined towards fibroblasts and AGS cells [62]. In a dose-response study, 32 reduced gastric lesions by 65% at 6 mg/kg and 31 by 54% at 100 mg/kg. The jatropholone B derivatives 33-38 and the compounds 39-42 were compared at a single oral dose of 25 mg/kg, while the jatropholone A derivatives 43-48 were assessed at 100 mg/kg. A decrease in gastroprotective activity was observed for the ether as well as for the ester derivatives of 32. The methyl and propyl ethers of 31 were more gastroprotective than the natural product The placement of an additional methyl group at C-2 in the jatropholone B derivatives led to a loss of selectivity; the methyl and propyl ethers lack a 2138 Natural Product Communications Vol. 3 (12) 2086 gastroprotective effect. At the dose of 25 mg/kg compound 32 reduced the lesions by 83%, while compound 31 inhibited them by only 36%. At 100 mg/kg, all the derivatives of 31 were active. Compounds 45-47 showed a similar activity to that of their parent 31, while derivatives 43, 44 and 48 were the most active. Considering the derivatives of 32, at 25 mg/kg, compounds 34, 41 and 42 showed the best gastroprotective effect, while compounds 38 and 39 were the less active. The gastroprotective mechanism of the natural diterpene ferruginol (49) was assessed in vivo. The involvement of gastric prostaglandins PGE(2), reduced GSH, NO or capsaicin receptors was evaluated in mice either treated or untreated with indomethacin, N-ethylmaleimide (NEM), N-nitro-Larginine methyl ester (L-NAME) or ruthenium red, respectively, and then orally treated with 49 or vehicle. Gastric lesions were induced by oral administration of ethanol. The effects of ferruginol (49) on the parameters of gastric secretion were assessed in pylorus-ligated rats. Gastric PGE(2) content was determined in rats treated with 49 and/or indomethacin. Tundis et al. OR1 OR H H H R O O H H 31 43 44 45 46 47 48 HO HO HO R=H R = CH3 R = C3H7 R = Ac R = COCH=CH2 R = COC6H4NO2p R = COC6H4Clp O OH O O HO RO OH OH H OH 32 33 34 35 36 37 38 39 40 41 42 OH O COOH O O O OH 49 Glc - O OH Glc - Glc - O H OH O O OH R = R1 = H R = H, R1 = CH3 R = H, R1 = C3H7 R = H, R1 = Ac R = H, R1 = COCH=CH2 R = H, R1 = COC6H4NO2p R = H, R1 = COC6H4Clp R = R1 = CH3 R = CH3, R1 = C3H7 R = C3H7, R1 = CH3, R = R1 = C3H7 52 50 R = H 51 R = Ac H OH stronger than those of the reference compounds, omeprazole and cimetidine [64]. The oligoglycoside fraction from the flower buds of Panax ginseng was found to show protective effects on ethanol-induced gastric mucosal lesions in rats. From this fraction, ginsenoside Rd (protopanaxadiol 3,20-O-bisdesmoside) (52) was isolated, together with new dammarane-type triterpene tetraglycosides. Ginsenoside Rd (52) exhibited inhibitory effects on ethanol- and indomethacin-induced gastric mucosal lesions in rats. The effect of 52 on ethanol-induced gastric lesions was equipollent to that of a reference compound, cetraxate hydrochloride [65]. The reduction of gastric GSH content was determined in rats treated with ethanol after oral administration of ferruginol (49), lansoprazole or vehicle. Finally, the acute oral toxicity was assessed in mice. Indomethacin reversed the gastroprotective effect of ferruginol (49) (25 mg/kg), but not NEM, ruthenium red or L-NAME. The diterpene (25 mg/kg) increased the gastric juice volume and its pH value, and reduced the titrable acidity, but was devoid of effect on the gastric mucus content. Ferruginol (49) increased gastric PGE(2) content in a dose-dependent manner and prevented the reduction in GSH observed due to ethanol-induced gastric lesions in rats. Single oral doses up to 3 g/kg 49 did not elicit mortality or acute toxic effects in mice. The results showed that ferruginol (49) acted as a gastroprotective agent stimulating gastric PGE(2) synthesis, reducing gastric acid output and improving the antioxidant capacity of the gastric mucosa by maintaining GSH levels [63]. The triterpene oleanolic acid (53) and its semisynthetic derivatives 54-59 were studied for gastroprotective and ulcer-healing effect using AGS cells and human lung fibroblasts (MRC-5) [66]. The assessment of the effect of the oleanolic acid derivatives on the PGE(2) content showed a significant increase of this prostaglandin when the AGS cell cultures were treated with compounds 53, 54, 56 and 58. The principal 28-noroleanane-type triterpene oligoglycosides camelliosides A (50) and B (51), isolated from the flowers buds of Camellia japonica, showed protective effects on both ethanol- and indomethacin-induced gastric lesions and their gastroprotective effects were either equivalent or The gastroprotective effect of oleanolic acid derivatives was assessed also in the HCl/EtOHinduced gastric lesions in mice. All the assayed compounds exhibited gastroprotective activity at the dose of 50 mg/kg, reducing the gastric lesions to different degrees ranging from 38% for compound 54 Gastroprotective natural products Natural Product Communications Vol. 3 (12) 2008 2139 R1 R1 H R = H, β OH; R1 = H2; R2 = H R = H, β Ac; R1 = H2; R2 = H R = H, β OH; R1 = H2; R2 = CH3 R = H, β Ac; R1 = H2; R2 = CH3 R = R1 = O; R2 = H O OH COOH O O O H OH O O HO O RO O OH OH OH H H OH OH HO O O OH O O 67 OH HO O O HO OH OR1 CH2OR2 OH R3 60 61 62 63 64 OH 65 R = OH 66 R = H H 58 R = R1 = O 59 R = H, β OH; R1 = H, α OH R HO HO O H R H 53 54 55 56 57 CO H OR2 H R O R O R = OAng, R1 = H, R2 = H, R3 = CH2OH R = OAng, R1 = H, R2 = Ac, R3 = CH2OH R = OAng, R1 = H, R2 = Ac, R3 = COOCH3 R = H, R1 = Ang, R2 = H, R3 = CHO R = OAng, R1 = Ac, R2 = H, R3 = CH2OH and up to 76% for compound 57. The most active products were compounds 57 and 59. In the compound group 53-56, differing in the free or esterified hydroxyl group at C-3 and the free or methylated carboxylic acid function at C-28, acetylation of the hydroxyl group at C-3 with a free COOH at C-28 reduced the gastroprotective activity, as can be observed for compound 54. Methylation of the COOH at C-28 in compound 57 significantly lowered the gastroprotective effect. Therefore, the effect should be related to the presence of a free carboxylic acid at C-28 when there is an oxo group at C-3 and C-11. In compounds 58 and 59, the free hydroxyl groups at C-3 and C-12 increased the gastroprotective effect, the activity of compound 58 being in the same range as that of oleanolic acid (53). The triterpene saponins theasaponins A1 (60), A2 (61), F3 (62), assamsaponin A (63) and assamsaponin D (64), isolated from the seeds of Camellia sinensis, were tested for their gastroprotective effects. Theasaponin A2 (61) showed an inhibitory effect on ethanol-induced gastric mucosal lesions in rats at a dose of 5.0 mg/kg, p.o. and its activity was more potent than that of omeprazole. Structure-activity relationships for theasaponins on ethanol-induced gastroprotective activities may suggest that (a) the 28-acetyl moiety enhances activity and (b) theasaponins having a 23-aldehyde group exhibit more potent activities than those with either a 23hydroxymethyl group or a 23-methoxycarbonyl group [67]. R O HO O R1 OCH 3 H 2 CO O O OCH 3 OCH 3 . H 2O OH OH O 71 68 R = OH, R1 = H 69 R = OH, R 1 = OH 70 R = OCH 3 , R 1 = OH 2.2. Flavonoids: The flavonoids minimiflorin (65) and mundulin (66) and the chalcone lonchocarpin (67), isolated from Lonchocarpus oaxacensis and L. guatemalensis, respectively, were tested on H+,K+ATPase isolated from dog stomach [68]. The flavanone minimiflorin (65) was the most potent inhibitor, while mundulin (66) was 7.3-fold less potent than 65. Hydroxylation at C-2' accounts for this difference in potency. Thus, hydroxylation plays an important role in conferring inhibitory activity of the gastric H+,K+-ATPase to the flavanones. Lonchocarpin (67), which has only one hydroxyl group in its molecule showed only moderate inhibition of ATPase (about 18-fold less potent than 65). A comparison of the relative potencies of these active compounds with omeprazole shows that many of these isolated compounds have higher inhibitory activity of H+,K+-ATPase than the reference compound, from 2 to 44 times higher for the most potent inhibitor of H+,K+-ATPase tested here, mundulin (66). Kolaviron is a mixture of three compounds, Garcinia biflavonoid GB1 (68), GB2 (69) and kolaflavanone (70) and has been extensively studied for its antiinflammatory property in various experimental models [69-72]. The antioxidant and scavenging properties of kolaviron have also been demonstrated [73]. Recently, it was demonstrated also that treatments with kolaviron significantly inhibited gastric lesions produced by indomethacin and acidified ethanol [74]. The effects of kolaviron on both indomethacin and ethanol-induced hemorrhagic erosion may be associated with an increase in gastric mucosal blood flow and gastric mucus secretion. 2140 Natural Product Communications Vol. 3 (12) 2086 OCH3O O O O CH2R N 72 R = H 73 R = OCH3 O N O 75 R = N NCH3 OH O R1 O R 79 R = 80 R = O O Some furoflavones (74-88), synthesized from the naturally occurring chromones visnagin (72) and khellin (73), exhibited gastroprotective activity in the ethanol damage model [77]. 81 R = H, R1 = C6H5 82 R = H, R1 = p-ClC6H4 R1 = C6H5 R1 = p-ClC6H4 83 R = 84 R = OCH3O N O R1 = C6H5 R1 = p-ClC6H4 OCH3O CH2R1 O R1 R 76 R = H, R1 = C6H5 77 R = H, R1 = p-ClC6H4 78 R = H, R1 = p-CH3C6H4 H2CN N 74 R = OCH3O O tablets to float in gastric fluid and release the drug continuously. The release of DA-6034 (71) from tablets in acidic media was significantly improved by using EFMS, which is attributed to the effect of the solubilizers and the alkalizing agent, such as sodium bicarbonate used as a gas generating agent. DA-6034 EFMS tablets showed enhanced gastroprotective effects in gastric ulcer-induced beagle dogs, indicating the therapeutic potential of EFMS tablets for the treatment of gastritis [76]. OCH3O O CH2R CH2 N O O R N O 87 R = OCH3 , R1 = NCH3 N O O 88 85 R = H, R1 = N(CH3)2 86 R = OCH3, R1 = N Tundis et al. The gastroprotective activity of DA-6034 (71), a new flavonoid derivative, against various ulcerogens including ethanol, aspirin, indomethacin, stress, and acetic acid was evaluated [75]. The basic mechanisms of DA-6034 (71) as a defensive factor, such as mucus secretion and endogenous PGE(2) synthesis were determined. Rats with gastric lesions induced by ethanol-HCl, aspirin, indomethacin, and stress that had been pretreated with 71 orally showed either a statistically significant decrease or decreasing tendency of the gastric lesion. In acetic acid-induced gastric lesions, repeated oral administration of 71 exhibited a U-shape activity in ulcer healing, with the maximum and minimum inhibition being observed at 30 and 10 mg/kg/day, respectively. DA-6034 (71) also increased the mucus content in the gel layer, as well as endogenous PGE(2) synthesis. These results suggest that 71 prevents gastric mucosal injury, and these gastroprotective activities appear to be due to the increase in the gastric defensive systems. The therapeutic limitations of 71 caused by its low solubility in acidic conditions were overcome by using the effervescent floating matrix system (EFMS), which was recently designed to cause In the benzopyrone portion of the furoflavone system, the type of aromatic substitution at the 7position affected the gastroprotective effect. The pmethoxyphenyl derivative 78 was more active than the p-chlorophenyl 77, which was more active than the phenyl derivative 76. The presence of a 9-alkylaminomethyl substituent in these compounds decreased the activity of 79 and 80, while the presence of a 6-alkylaminomethyl substituent increased the activity (compound 86 showed more activity than 76). When the aromatic group in position 7 was pyridinyl, the activity was slightly decreased (compounds 74 and 85), except in the case of the 9-N-methylpiperazinomethyl derivative 75, which showed promising gastroprotective activity. It was found that the presence of a methoxy group showed great effect on the activity. Substitution at the 4-position with a methoxy group (compounds 76-80) enhanced the gastroprotective activity, in contrast to 4-hydroxy derivatives 81-83, which showed a marked decrease in activity. Substitution with another methoxy group (87) produced a potent level of gastroprotection. In summary, furoflavones exhibited good gastroprotective activity in the ethanol damage model when there was a methoxy group (either in the 4, 9 or 7-position as methoxyphenyl) and an appropriate substitution in the 6-position with an alkylaminomethyl group. 2.3. Xanthones: Four xanthones, 6-desoxyjacareubin (89), jacareubin (90), 1,3,5,6-tetra-hydroxy-2-(3hydroxy-3-methylbutyl)-xanthone (91) and 1- Gastroprotective natural products O R OH O OH Natural Product Communications Vol. 3 (12) 2008 2141 O O HO O OH 91 OH O O OAc HO OH O OH OAc 92 content in mice, suggesting an antioxidant action. These findings provide evidence that mangiferin (93) affords gastroprotection against gastric injury induced by ethanol and indomethacin, most possibly through the antisecretory and antioxidant mechanisms of action [78]. β-D-glucopyranosyl HO AcO OH OH 89 R = H 90 R = OH O OH 93 hydroxy-3,5,6-tri-O-acetyl-2(3,3-dimethylallyl) xanthone (92), isolated from Calophyllum brasilienses were tested on H+,K+-ATPase isolated from dog stomach [68]. The compounds showed IC50values ranging from 47 μM to 1.6 mM. Steric hindrance by the substituents at C-6 and C-3 appears to influence the potency of inhibition of H+,K+-ATPase activity of these compounds. The presence of a hydroxyl group at C-6 seems to play a prime role in the activity of xanthones on gastric ATPase. In accord with this, groups at C-6 in xanthone 89 reduced the potency of H+,K+-ATPase inhibition by 34-fold. In addition, acetylation at this position (xanthone 91) also reduced the activity of the enzyme by a similar amount. Also, the presence of a bulky substituent at C-3 significantly reduced the potency of inhibition of gastric H+,K+-ATPase activity by xanthone 91. In search of novel gastroprotective agents, mangiferin (93), a naturally occurring glucosylxanthone from Mangifera indica, was evaluated in mice suffering gastric injury induced by ethanol and indomethacin. The effects of 93 on gastric mucosal damage were assessed by determination of changes in either mean gastric lesion area or ulcer score in mice and on gastric secretory volume and total acidity in 4 hour pylorus-ligated rats. Mangiferin (93) (3, 10 and 30 mg/kg p.o.) significantly attenuated the gastric damage induced by ethanol and indomethacin. NAcetylcysteine (750 mg/kg, i.p.) and lansoprazole (30 mg/kg, p.o.), used as positive controls in these ulcerogenic models, resulted in 50% and 76% suppression of gastric injury, respectively. In 4 hourpylorus-ligated rats, intraduodenally applied 93 (30 mg/kg) caused significant diminutions in gastric secretory volume and total acidity. In addition, like N-acetylcysteine, a donor of sulfhydryls, mangiferin (93) effectively prevented the ethanol-associated depletion of gastric mucosal non-protein sulfhydryl Conclusions: The development of safe and effective drugs capable of preventing stomach damage induced by NSAIDs or other gastric-damaging substances represents an important goal of medicinal research considering the large use of these drugs and the increased healthcare costs when peptic ulcer disease becomes a chronic condition. It is well established that natural products are an excellent source of chemical structures with a wide variety of biological activity, including gastroprotective properties. The large number of compounds derived from natural sources that are currently undergoing evaluation in clinical trials is another positive indicator that natural product discovery provides good value for human medicine. This paper gives an up-to-date review of plant extracts, natural compounds and their derivatives as gastroprotective agents. This knowledge should encourage further in vitro and in vivo pharmacological studies and help to provide leads to the ultimate goal of developing novel gastroprotective drugs. List of abbreviations AGS CAT COX-2 CYP1A1 EFMS GPx GSH IC50 IL-8 iNOS LD50 L-NAME LPO MRC-5 NEM NF-κB NO NP-SH NSAIDs PGE ROS SOD XO = Human gastric epithelial cells = Catalase = Cyclooxygenase-2 = Cytochrome P450 1A1 = Effervescent floating matrix system = Glutathione peroxidase = Glutathione = Inhibitory concentration 50% = Interleukin-8 = Inducible nitric oxide synthase = Lethal dose 50% = N-nitro-L-arginine methyl ester = Lipid peroxidation = Human lung fibroblasts = N-ethylmaleimide = Nuclear factor kappa B = Nitric oxide = Non-protein sulfhydryl = Nonsteroidal anti-inflammatory drugs = Prostaglandin = Reactive oxygen species = Superoxide dismutase = Xanthine oxidase 2142 Natural Product Communications Vol. 3 (12) 2086 Tundis et al. References [1] [2] [3] [4] [5] [6] [7] [8] [9] [10] [11] [12] [13] [14] [15] [16] [17] [18] [19] [20] [21] [22] [23] [24] [25] [26] [27] [28] Kurata JH, Nogawa AN. (1997) Meta-analysis of risk factors for peptic ulcer. Nonsteroidal anti-inflammatory drugs, Helicobacter pylori, and smoking. Journal of Clinical Gastroenterology, 24, 2-17. Nilsson C, Sillen A, Eriksson L, Strand ML, Enroth H, Normark S, Falk P, Engstrand L. (2003) Correlation between cag pathogenicity island composition and Helicobacter pylori-associated gastroduodenal disease. Infection and Immunity, 71, 65736581. Wallace JL, Granger DN. (1992) Pathogenesis of NSAID gastropathy: are neutrophils the culprits? Trends in Pharmacological Sciences, 13, 129-131. Wolfe MM, Sachs G. (2000) Acid suppression: optimizing therapy for gastroduodenal ulcer healing, gastroesophageal reflux disease, and stress-related erosive syndrome. Gastroenterology, 118, S9-S31. WHO. (2003) WHO Traditional Medicine Fact Sheet No. 134. WHO: Geneva. Kuruvilla A. (2002) Herbal formulations as pharmacotherapeutic agents. Indian Journal of Experimental Biology, 40, 7-11. Gracioso JS, Hiruma-Lima CA, Souza Brito AR. (2000) Antiulcerogenic effect of a hydroalcoholic extract and its organic fractions of Neurolaena lobata (L.) R.Br. Phytomedicine, 7, 283-289. Hiruma-Lima CA, Gracioso JS, Bighetti EJ, Grassi-Kassisse DM, Nunes DS, Brito AR. (2002) Effect of essential oil obtained from Croton cajucara Benth. on gastric ulcer healing and protective factors of the gastric mucosa. Phytomedicine, 9, 523-529. Toma W, Gracioso JS, Hiruma-Lima CA, Andrade FD, Vilegas W, Souza Brito ARM. (2003) Evaluation of the analgesic and antiedematogenic activities of Quassia amara bark extract. Journal of Ethnopharmacology, 85, 19-23. Schmeda-Hirschmann G, Yesilada E. (2005) Traditional medicine and gastroprotective crude drugs. Journal of Ethnopharmacology, 100, 61-66. de Andrade SF, Lemos M, Comunello E, Noldin VF, Filho VC, Niero R. (2007) Evaluation of the antiulcerogenic activity of Maytenus robusta (Celastraceae) in different experimental ulcer models. Journal of Ethnopharmacology, 113, 252-257. Naik Y, Jayaram S, Harish Nayaka MA, Lakshman , Dharmesh SM. (2007) Gastroprotective effect of swallow root (Decalepis hamiltonii) extract: possible involvement of H(+)-K(+)-ATPase inhibition and antioxidative mechanism. Journal of Ethnopharmacology, 112, 173-179. Berenguer B, Trabadela C, Sánchez-Fidalgo S, Quílez A, Miño P, De la Puerta R, Martín-Calero MJ. (2007) The aerial parts of Guazuma ulmifolia Lam. protect against NSAID-induced gastric lesions Journal of Ethnopharmacology, 114, 153-160. Lapa F da R, Freitas CS, Baggio CH, Missau FC, Pizzolatti MG, Santos AR, Marques MC. (2007) Gastroprotective activity of the hydroalcoholic extract obtained from Polygala paniculata L. in rats. Journal of Pharmacy and Pharmacology, 59, 1413-1419. Devi RS, Narayan S, Vani G, Srinivasan P, Mohan KV, Sabitha KE, Devi CS. (2007) Ulcer protective effect of Terminalia arjuna on gastric mucosal defensive mechanism in experimental rats. Phytotherapy Research, 21, 762-767. Al-Mofleh IA, Alhaider AA, Mossa JS, Al-Sohaibani MO, Rafatullah S, Qureshi S. (2005) Inhibition of gastric mucosal damage by Piper nigrum (black pepper). Pharmacognosy Magazine, 1, 64-68. Rafatullah S, Tariq M, Al-Yahya MA, Mossa JS, Ageel AM. (1990) Evaluation of turmeric (Curcuma longa) for gastric and duodenal antiulcer activity in rats. Journal of Ethnopharmacology, 29, 25-34. al-Yahya MA, Rafatullah S, Mossa JS, Ageel AM, Parmar NS, Tariq M. (1989) Gastroprotective activity of ginger Zingiber officinale Rosc., in albino rats. The American Journal of Chinese Medicine, 17, 51-56. Al Mofleh IA, Alhaider AA, Mossa JS, Al-Soohaibani MO, Rafatullah S. (2007) Aqueous suspension of anise "Pimpinella anisum" protects rats against chemically induced gastric ulcers. World Journal of Gastroenterology, 13, 1112-1118. Al-Mofleh IA, Alhaider AA, Mossa JS, Al-Sohaibani MO, Qureshi S, Rafatullah S. (2006) Antisecretagogue, antiulcer and cytoprotective effects of ‘peppermint’ Mentha piperita L. in laboratory animals. Journal of Medical Sciences, 6, 930-936. Alhaider AA, Al-Mofleh IA, Al-Sohaibani MO, Rafatullah S, Qureshi S. (2006) Effect of Carum carvi on experimentally induced gastric mucosal damage in Wistar albino rats. International Journal of Pharmacology, 2, 309-315. Al-Mofleh IA, Alhaider AA, Mossa JS, Al-Sohaibani MO, Qureshi S, Rafatullah S. (2006) Protection of gastric mucosal damage by Coriandrum sativum L. pretreatment in Wistar albino rats. Environmental Toxicology and Pharmacology, 22, 64-69. Al-Mofleh IA, Alhaider AA, Mossa JS, Al-Sohaibani MO, Qureshi S, Rafatullah S. (2006). Antigastric ulcer studies on “saffron” Crocus sativus L. in rats. Pakistan Journal of Biological Sciences, 9, 1009-1013. Dordević S, Petrović S, Dobrić S, Milenković M, Vucićević D, Zizić S, Kukić J. (2007) Antimicrobial, anti-inflammatory, anti-ulcer and antioxidant activities of Carlina acanthifolia root essential oil. Journal of Ethnopharmacology, 109, 458-463. Terano A, Hiraishi H, Ota S, Shi Tega J, Sugimoto T. (1989) Role of superoxide and hydroxyl radicals in rat gastric mucosal injury induced by ethanol. Gastroenterologia Japonica, 24, 488-493. Shih YT, Wu DC, Liu CM, Yang YC, Chen IJ, Lo YC. (2007) San-Huang-Xie-Xin-Tang inhibits Helicobacter pylori-induced inflammation in human gastric epithelial AGS cells. Journal of Ethnopharmacology, 112, 537-544. Chu SH, Kim H, Seo JY, Lim JW, Mukaida N, Kim KH. (2003) Role of NF-kappaB and AP-1 on Helicobacter pylori-induced IL-8 expression in AGS cells. Digestive Diseases and Sciences, 48, 257-265. Kim H, Seo JY, Kim KH. (2002) Effect of mannitol on Helicobacter pylori-induced cyclooxygenase-2 expression in gastric epithelial AGS cells. Pharmacology, 66, 182-189. Gastroprotective natural products [29] [30] [31] [32] [33] [34] [35] [36] [37] [38] [39] [40] [41] [42] [43] [44] [45] [46] [47] [48] [49] [50] [51] [52] [53] [54] [55] Natural Product Communications Vol. 3 (12) 2008 2143 Walker MM. (2002) Cyclooxygenase-2 expression in early gastric cancer, intestinal metaplasia and Helicobacter pylori infection. European Journal of Gastroenterology & Hepatology, 14, 347-349. Kim H, Lim JW, Kim KH. (2001) Helicobacter pylori-induced expression of interleukin-8 and cyclooxygenase-2 in AGS gastric epithelial cells: mediation by nuclear factor-kappaB. Scandinavian Journal of Gastroenterology, 36, 706-716. Chang YJ, Wu MS, Lin JT, Sheu BS, Muta T, Inoue H, Chen CC. (2004) Induction of cyclooxygenase-2 over expression in human gastric epithelial cells by Helicobacter pylori involves TLR2/TLR9 and c-Srcdependent nuclear factor-kappaB activation. Molecular Pharmacology, 66, 1465-1477. Harris PR, Mobley HL, Perez-Perez GI, Blaser MJ, Smith PD. (1996) Helicobacter pylori urease is a potent stimulus of mononuclear phagocyte activation and inflammatory cytokine production. Gastroenterology, 111, 419-425. Calvo TR, Lima ZP, Silva JS, Ballesteros KV, Pellizzon CH, Hiruma-Lima CA, Tamashiro J, Brito AR, Takahira RK, Vilegas W. (2007) Constituents and antiulcer effect of Alchornea glandulosa: activation of cell proliferation in gastric mucosa during the healing process. Biological & Pharmaceutical Bulletin, 30, 451-459. Govindarajan R, Vijayakumar M, Singh M, Rao ChV, Shirwaikar A, Rawat AK, Pushpangadan P. (2006) Antiulcer and antimicrobial activity of Anogeissus latifolia. Journal of Ethnopharmacology, 106, 57-61. Rao ChV, Sairam K, Goel RK. (2000) Experimental evaluation of Bacopa monniera on rat gastric ulceration and secretion. Indian Journal of Physiology and Pharmacology, 44, 35-41. Sairam K, Rao ChV, Babu DM, Goel RK. (2002) Antiulcerogenic activity of methanolic extract of Emblica officinalis. Journal of Ethnopharmacology, 82, 1-9. Hase T, Moss BJ. (1973) Microvascular changes of gastric mucosa in development of stress ulcers in rats. Gastroenterology, 65, 224-226. Cho CH, Ogle CW. (1979) Cholinergic-mediated gastric mast cell degranulation with subsequent histamine H1 and H2-receptor activation in stress ulceration in rats. European Journal of Pharmacology, 55, 23-33. Rao ChV, Maiti RN, Goel RK. (1999) Effect of mild irritant on gastric mucosal offensive and defensive factors. Indian Journal of Physiology and Pharmacology, 44, 185-191. Penido C, Costa KA, Futuro DO, Paiva SR, Kaplan MA, Figueiredo MR, Henriques MG. (2006) Anti-inflammatory and antiulcerogenic properties of Stachytarpheta cayennensis (L.C. Rich) Vahl. Journal of Ethnopharmacology, 104, 225-233. Andreo MA, Ballesteros KV, Hiruma-Lima CA, Machado da Rocha LR, Souza Brito AR, Vilegas W. (2006) Effect of Mouriri pusa extracts on experimentally induced gastric lesions in rodents: role of endogenous sulfhydryl compounds and nitric oxide in gastroprotection. Journal of Ethnopharmacology, 107, 431-441. Berenguer B, Sánchez LM, Quílez A, López-Barreiro M, de Haro O, Gálvez J, Martín MJ. (2006) Protective and antioxidant effects of Rhizophora mangle L. against NSAID-induced gastric ulcers Journal of Ethnopharmacology, 103, 194-200. Sánchez Perera LM, Ruedas D, Gómez BC. (2001) Gastric antiulcer effect of Rhizophora mangle L. Journal of Ethnopharmacology, 77, 1-3. Gonzales E, Iglesias I, Carretero E, Villar A. (2000) Gastric cytoprotection of Bolivian medicinal plants. Journal of Ethnopharmacology, 70, 329-333. Tebid K, Besancon P, Rovanet JM. (1996) Effects of dietary grape seed tannins on rat cecal fermentation and colonic bacterial enzymes. Nutrition Research, 16, 105-110. Ramírez RO, Roa CC. (2003) The gastroprotective effect of tannins extracted from duhat (Syzygium cumini Skeels) bark on HCl/ethanol induced gastric mucosal injury in Sprague-Dawley rats. Clinical Hemorheology and Microcirculation, 29, 253-261. Fernandez O, Capdevila JZ, Dalla G, Melchor G. (2002) Efficacy of Rhizophora mangle aqueous bark extract in the healing of open surgical wounds. Fitoterapia, 73, 564-568. Jamal A, Javed K, Aslam M, Jafri MA. (2006) Gastroprotective effect of cardamom, Elettaria cardamomum Maton. fruits in rats. Journal of Ethnopharmacology, 103, 149-153. Rujjanawate C, Kanjanapothi D, Amornlerdpison D, Pojanagaroon S. (2005) Anti-gastric ulcer effect of Kaempferia parviflora. Journal of Ethnopharmacology, 102, 120-122. Sannomiya M, Fonseca VB, da Silva MA, Rocha LR, Dos Santos LC, Hiruma-Lima CA, Souza Brito AR, Vilegas W. (2005) Flavonoids and antiulcerogenic activity from Byrsonima crassa leaves extracts. Journal of Ethnopharmacology, 97, 1-6. Kushima H, Hiruma-Lima CA, Santos MA, Viana E, Coelho-Ferreira M, Brito AR. (2005) Gastroprotective activity of Pradosia huberi on experimentally induced gastric lesions in rodents: role of endogenous sulphydryls and nitric oxide. Journal of Ethnopharmacology, 101, 61-67. Shan L, Liu RH, Shen YH, Zhang WD, Zhang C, Wu DZ, Min L, Su J, Xu XK. (2006) Gastroprotective effect of a traditional Chinese herbal drug "Baishouwu" on experimental gastric lesions in rats. Journal of Ethnopharmacology, 107, 389-394. Jainu M, Mohan KV, Devi CS. (2006) Protective effect of Cissus quadrangularis on neutrophil mediated tissue injury induced by aspirin in rats. Journal of Ethnopharmacology, 104, 302-305. Ajaikumar KB, Asheef M, Babu BH, Padikkala J. (2005) The inhibition of gastric mucosal injury by Punica granatum L. (pomegranate) methanolic extract. Journal of Ethnopharmacology, 96, 171-176. Al-Qarawi AA, Abdel-Rahman H, Ali BH, Mousa HM, El-Mougy SA. (2005) The ameliorative effect of dates (Phoenix dactylifera L.) on ethanol-induced gastric ulcer in rats. Journal of Ethnopharmacology, 98, 313-317. 2144 Natural Product Communications Vol. 3 (12) 2086 [56] [57] [58] [59] [60] [61] [62] [63] [64] [65] [66] [69] [67] [68] [69] [70] [71] [72] [73] [74] [75] [76] [77] [78] Tundis et al. Karimi G, Hosseinzadeh H, Ettehad N. (2004) Evaluation of the gastric antiulcerogenic effects of Portulaca oleracea L. extracts in mice. Phytotherapy Research, 18, 484-487. Barocelli E, Calcina F, Chiavarini M, Impicciatore M, Bruni R, Bianchi A, Ballabeni V. (2004) Antinociceptive and gastroprotective effects of inhaled and orally administered Lavandula hybrida Reverchon “Grosso” essential oil. Life Sciences, 76, 213-223. Pertino M, Schmeda-Hirschmann G, Rodríguez JA, Theoduloz C. (2007) Gastroprotective effect and cytotoxicity of terpenes from the Paraguayan crude drug “yagua rova” (Jatropha isabelli). Journal of Ethnopharmacology, 22, 553-559. Pertino M, Rodríguez JA, Theoduloz C, Razmilic I, Schmeda-Hirschmann G. (2006) Gastroprotective activity and cytotoxic effect of cyperenoic acid derivatives. Journal of Pharmacy and Pharmacology, 58, 1507-1513. Reyes M, Schmeda-Hirschmann G, Razmilic I, Theoduloz C, Yáñez T, Rodríguez JA. (2005) Gastroprotective activity of sesquiterpene derivatives from Fabiana imbricata. Phytotherapy Research, 19, 1038-1042. Izquierdo R, Astudillo L, Rodríguez JA, Theoduloz C, Palenzuela JA, Schmeda-Hirschmann G. (2007) Gastroprotective effect and cytotoxicity of labdenamides. Planta Medica, 73, 310-317. Pertino M, Schmeda-Hirschmann G, Rodríguez JA, Theoduloz C. (2007) Gastroprotective effect and cytotoxicity of semisynthetic jatropholone derivatives. Planta Medica, 73, 1095-100. Areche C, Theoduloz C, Yáñez T, Souza-Brito AR, Barbastefano V, de Paula D, Ferreira AL, Schmeda-Hirschmann G, Rodríguez JA. (2008) Gastroprotective activity of ferruginol in mice and rats: effects on gastric secretion, endogenous prostaglandins and nonprotein sulfhydryls. Journal of Pharmacy and Pharmacology, 60, 245-251. Yoshikawa M, Morikawa T, Asao Y, Fujiwara E, Nakamura S, Matsuda H. (2007) Medicinal flowers. XV. The structures of noroleanane- and oleanane-type triterpene oligoglycosides with gastroprotective and platelet aggregation activities from flowers buds of Camellia japonica. Chemical & Pharmaceutical Bulletin, 55, 606-612. Yoshikawa M, Sugimoto S, Nakamura S, Sakumae H, Matsuda H. (2007) Medicinal flowers. XVI. New dammarane-type triterpene tetraglycosides and gastroprotective principles from flower buds of Panax ginseng. Chemical & Pharmaceutical Bulletin, 55, 1034-1038. Sánchez M, Theoduloz C, Schmeda-Hirschmann G, Razmilic I, Yáñez T, Rodríguez JA. (2006) Gastroprotective and ulcer-healing activity of oleanolic acid derivatives: in vitro-in vivo relationships. Life Sciences, 79, 1349-1356. Morikawa T, Li N, Nagatomo A, Matsuda H, Li X, Yoshikawa M. (2006) Triterpene saponins with gastroprotective effects from tea seed (the seeds of Camellia sinensis). Journal of Natural Products, 69, 185-190. Reyes-Chilpa R, Baggio CH, Alavez-Solano D, Estrada-Muñiz E, Kauffman FC, Sanchez RI, Mesia-Vela S. (2006) Inhibition of gastric H+,K+-ATPase activity by flavonoids, coumarins and xanthones isolated from Mexican medicinal plants. Journal of Ethnopharmacology, 105, 167-172. Akintonwa A, Essien AR. (1990) Protective effects of Garcinia kola seed extract against paracetamol-induced hepatotoxicity in rats. Journal of Ethnopharmacology, 29, 207-211. Olaleye SB, Farombi EO, Adewoye EO, Onasanwo SO, Owoyele BV, Elegbe RA. (2000) Analgesic and anti-inflammatory effects of kolaviron (a Garcinia kola seed extract). African Journal of Biomedical Research, 3, 163-166. Farombi EO, Tahnteng JG, Agboola O, Nwankwo JO, Emerole GO. (2000) Chemoprevention of 2-acetyl aminofluoreneinduced hepatotoxicity and lipid peroxidation in rats by kolaviron - a Garcinia kola seed extract. Food and Chemical Toxicology, 38, 535-541. Farombi EO, Adepoju BF, Ola-Davies OE, Emerole GO. (2001) Inhibition of aflatoxin B1-induced clastogenicity and hepatocarcinogenicity by kolaviron (Garcinia biflavanones) in rats. Mutation Research, 483, S106. Farombi EO, Akanni OO, Emerole GO. (2002) Antioxidative and scavenging activities of kolaviron in vitro. Pharmaceutical Biology, 40, 107-116. Farombi EO, Akuru TO, Alabi MC. (2002) Kolaviron modulates cellular redox status and impairment of membrane protein activities by potassium bromate (KBrO3) in rats. Pharmacological Research, 45, 63-68. Olaleye SB, Farombi EO. (2006) Attenuation of indomethacin- and HCl/ethanol-induced oxidative gastric mucosa damage in rats by kolaviron, a natural biflavonoid of Garcinia kola seed. Phytotherapy Research, 20, 14-20. Choi SM, Shin JH, Kang KK, Ahn BO, Yoo M. (2007) Gastroprotective effects of DA-6034, a new flavonoid derivative, in various gastric mucosal damage models. Digestive Diseases Sciences, 52, 3075-3080. Jang SW, Lee JW, Park SH, Kim JH, Yoo M, Na DH, Lee KC. (2008) Gastroretentive drug delivery system of DA-6034, a new flavonoid derivative, for the treatment of gastritis. International Journal of Pharmaceutics, 356, 88-94. Ragab FA, Hassan GS, Yossef HA, Hashem HA. (2007) Synthesis of 6- and 9-alkylaminomethyl furoflavones as gastroprotective agents. European Journal of Medicinal Chemistry, 42, 1117-1127. Carvalho AC, Guedes MM, de Souza AL, Trevisan MT, Lima AF, Santos FA, Rao VS. (2007) Gastroprotective effect of mangiferin, a xanthonoid from Mangifera indica, against gastric injury induced by ethanol and indomethacin in rodents. Planta Medica, 73, 1372-1376. NPC Natural Product Communications Phytochemistry and Pharmacology of Boronia pinnata Sm. 2008 Vol. 3 No. 12 2145 - 2150 MassimoCurinia*, Salvatore Genovesea, Luigi Menghinib, Maria Carla Marcotullioa and Francesco Epifanob a Dipartimento di Chimica e Tecnologia del Farmaco, Sezione di Chimica Organica, Università degli Studi di Perugia, Via del Liceo, Perugia, Italy 06123 b Dipartimento di Scienze del Farmaco, Università “G. D’Annunzio”, Via dei Vestini 31, 66013, Chieti Scalo (CH), Italy 66013 curmax@unipg.it Received: July 1st, 2008; Accepted: October 21st, 2008 Boronia pinnata Sm. (Rutaceae) is a plant that is widespread in New South Wales (Australia). Although there are no reports about the use of this species in the local ethnomedical traditions, recent investigations led to the characterization of several secondary metabolites, most belonging to the class of prenyloxyphenylpropanoids. Some of the compounds extracted from B. pinnata showed valuable biological properties, such as anti-inflammatory activity and in vitro inhibition of growth of Helicobacter pylori. The aim of this review is to cover what has been reported so far in the literature on the title plant from a phytochemical and pharmacological point of view. Keywords: Anti-inflammatory activity, Boronia pinnata, Helicobacter pylori, prenyloxyphenylpropanoids. Prenylation is a chemical or enzymatic addition of an hydrophobic side chain to an accepting molecule (another terpenoid molecule, an aromatic compound, a protein, etc.). In particular, prenylation of aromatic secondary metabolites plays a critical role in the biosynthesis of a wide range of molecules exerting valuable pharmacological effects across phylogenetically different classes of living organisms, from bacteria to mammals and plants. Frequently, the addition of an isoprenoid chain renders the molecule more effective than the parent compound from a pharmacological point of view. These ‘‘hybrid’’ natural products represent nowadays a new frontier for the development of novel drugs, in particular as antimicrobial, anti-oxidant, antiinflammatory and anti-cancer agents. Oxyprenylated natural products are compounds of mixed biosynthetic origin for which the final step of the biosynthetic process is the prenylation of either an alkaloid or a phenylpropanoid core using prenyl diphosphate as alkylating agent [1], the latter coming in turn from either the mevalonate [2] or 1-DOXP pathways [3]. Oxyprenylated secondary metabolites have been considered for decades merely as biosynthetic intermediates of C-prenylated compounds and only in the last ten years have been characterized as phytochemicals exerting interesting and valuable biological activities [4]. Considering the length of the carbon chain, three types of prenyloxy skeletons can be identified: C5 (isopentenyl), C10 (geranyl) and C15 (farnesyl). Isopentenyloxy and geranyloxy chains are quite abundant in nature, while farnesyloxy ones are less common. The skeleton may consist only of carbon and hydrogen or may contain oxygen atoms, usually in the form of alcohols, ethers or ketones. Several species have been identified up to now as producing prenyloxyphenylpropanoids. Among these Boronia pinnata Sm. (Rutaceae) has been characterized as one of the species to biosynthesize a wide variety of the above cited secondary metabolites. While several plants of the genus Boronia have been reported to be used in local ethnomedical traditions [5-10], B. pinnata has never been cited in the literature in this regard. However, recent investigations led to a detailed phytochemical profile of the main secondary metabolites of this plant and have revealed that some of these compounds exert valuable anti-cancer and anti-inflammatory effects, 2146 Natural Product Communications Vol. 3 (12) 2008 mainly targeting the lipoxygenase system, and an inhibitory activity in vitro against Helicobacter pylori. The aim of this review is to cover what has been reported so far in the literature on the title plant from a phytochemical and pharmacological point of view. Curini et al. OH CH3O HO O OH CH3O OGlcRhm OCH3 OH 1 O 2 R1 Botany of Boronia pinnata B. pinnata is a species belonging to the family Rutaceae that typically grows in a very restricted area of Australia, namely New South Wales. The name of the genus comes from Francesco Borone, an Italian botanist (1769-1794), while “pinnata” alludes to the paired leafets that are an anatomical feature of this plant [10]. B. pinnata grows in sandstone country up to lower mountain levels in dry sclerophyll forests and in well-drained sandy heaths, mainly along coasts. It is a beautiful waxy-flowered shrub, 0.5-1.5 m high. Branchlets are glabrous and slightly angled to prominently 4-angled. The foliage is ornamental and ferny, light to mid-green. Leaves are up to 25 mm long, opposite and with several pairs of widely spaced leaflets. Flowers are grouped in inflorescences that are subterminal to axillary, collected in corymbose cymes, each comprising from 3 to 8 flowers. Petals are in group of 4, imbricate, 5-10 mm long, colored bright to purplish pink. The flowering stage occurs from June to February [11]. R2 R4 OR3 3 R1 CHO 4 CHO R2 R3 H R4 H H H O 5 CHO OCH3 H 6 CHO H H 7 8 9 CHO CHO CH2OH OCH3 OCH3 OCH3 CH3 CH3 H OCH3 H 10 11 CH2OCH3 CH2OH OCH3 OCH3 CH3 OCH3 OCH3 12 13 14 15 CH2OH CH2OH COOH COOH OCH3 OCH3 H OCH3 CH3 CH3 H H OCH3 H H Phytochemistry of B. pinnata The first studies describing the phytochemical composition of B. pinnata refer to the analysis of the essential oil obtained from the aerial parts of the plant, which were reported in two manuscripts published in 1919 and 1922 [12,13]. The essential oil was obtained with a yield of 0.1% and showed a very high quantity of elemicin (1) (> 75%) that was also claimed as a distinctive feature of B. pinnata in comparison with other Boronia species. At the same time, Morrison reported the isolation of rutin (2) from the aqueous extract of the leaves [14]. R1 R3 OR2 R1 H R2 16 R3 OCH3 17 OCH3 CH3 H OCH3 O O H OH H OH O Studies on the title plant were then abandoned for about 80 years. In 1999 and 2000 Itoigawa et al reported the isolation and structural characterization of 23 novel and already known secondary metabolitesfrom the roots of B. pinnata [15,16]. In particular, they isolated six cinnamic aldehyde derivatives, boropinals A (3), B (4) and C (5), together with 3’,4’-dimethoxycinnamic aldehyde (7) and 3’,4’,5’-trimethoxycinnamic aldehyde (8), five cinnamoyl alcohol derivatives, boropinol A (9), OH N OCH3 O CH3 18 19 OCH3 R1 R2 N O R3 20 21 R1 H H R2 H OCH3 R3 H H Phytochemical and pharmacological aspects of Boronia pinnata Natural Product Communications Vol. 3 (12) 2008 2147 OCH3 1 N OCH3 H R a O HO O 25 R1 OCH3 R2 23 H H 15 CH3O O OCH3 OCH3 22 O CHO CHO R2 methoxyboropinol B (10), boropinol C (11), 3’,4’dimethoxycinnamyl alcohol (12) and 3’,4’,5’trimethoxycinnamyl alcohol (13), two cinnamic acids, p-hydroxycinnamic (14) and boropinic acid (15), three phenylpropenes, 3-(3’-methoxy-4’prenyloxy)phenyl-1-propene (16), methyleugenol (17) and elemicin (1), one lignan, boropinan (18), five alkaloids, pinolinone (19), dictamnine (20), evolitrine (21), preskimmianine (22) and folimine (23), and finally one coumarin, braylin (24). Some of the novel secondary metabolites extracted from the roots of B. pinnata have been also obtained by chemical synthesis. The first synthesis in this regard was reported by Hou and coworkers in 2003 [9]. They synthesized boropinol A (9), boropinal C (5) and boropinic acid (15) using commercially available vanillin (25) as starting material (Scheme 1). Condensation of the latter with α-carbethoxy-methylphosphorane in DME furnished the corresponding ester, followed by alkaline hydrolysis to give boropinic acid (15) in 91% yield. Reduction of 15 with LiAlH4 in Et2O yielded boropinol A (9), although not in a sufficiently pure form. So the mixture was oxidized with K2Cr2O7 in DMSO to give boropinal C (5) in 61% yield. Finally, reduction of 5 with NaBH4 in MeOH yielded pure boropinol A (9) (70%) [17]. The reaction of 25 with isopentenyl bromide in DMF gave the O-prenylated adduct 26 in 84% yield. Three years later Curini and coworkers reported a short and high-yielding synthesis of boropinic acid (15) starting from readily commercially available ferulic acid (7) (Scheme 2) [18]. d,e 5 26 f 9 Scheme 1: Reagents and conditions: (a) isopentenyl bromide, DMF; (b) Ph3P=CH-CO2Et, DME; (c) KOH (aq); (d) LAH, dry Et2O; (e) K2Cr2O7, DMSO; (f) NaBH4, MeOH. CO2H O 24 b,c CO2CH3 a HO HO OCH3 OCH3 7 b,c 28 15 Scheme 2: Reagents and conditions: (a) MeOH, conc. H2SO4, reflux; (b) isopentenyl bromide, K2CO3, acetone, reflux; (c) NaOH 70°C. Ferulic acid (7) was first converted to the corresponding methyl esters 28 in 99% yield by reaction in refluxing MeOH under the catalysis of conc. H2SO4; compound 28 was submitted to prenylation using isopentenyl bromide as alkylating agent and dry K2CO3 as base in refluxing acetone, followed by alkaline hydrolysis in the same reaction vessel to obtain boropinic acid (16) in 96% yield. So, compound 16 has been easily synthesized from widely available and non-toxic starting materials by a high-yielding, environment friendly, and cheap synthetic route. Pharmacological properties of active principles from B. pinnata The fact that B. pinnata was not part of local ethnomedical traditions, mostly due to its restricted habitat, has not attracted, for many decades, the attention of researchers to carry out pharmacological studies of extracts or single secondary metabolites of this plant. The first study, aimed at investigating the anticancer properties of selected compounds extracted from roots of B. pinnata, was reported by Itoigawa and coworkers in 1999 [15]. Twelve phenylpropanoids, namely compounds 1, 3, 5, 7-12, 16 and 17 were tested in vitro as inhibitors of Epstein-Barr virus early antigen (EBV-EA) activation induced by 12-O-tetradecanoylphorbol-13-acetate (TPA) in Raji cells (EBV genome-carrying human lymphoblastoid cells; EBV non –producer type). One of the extracted compounds, 3-(3’-methoxy-4’isopentenyloxy)phenyl-1-propene (16), was also tested in vivo as an inhibitor of effects on skin tumor 2148 Natural Product Communications Vol. 3 (12) 2008 Curini et al. Table 1: Inhibitory effects of selected phenylpropanoids from roots of B. pinnata on TPA-induced EBV-EA activation. Table 2: Inhibitory effects of compounds 3 and 14 on TPA-induced EBV-EA activation. Compd. Compd. 1 3 5 7 8 9 10 11 12 13 16 17 EBV-EA positive cells (% vialibility) Compd. concentration (mol ratio/32 pmol TPA) 500 100 10 29.2 ± 1.6 71.2 ± 2.2 90.1 ± 1.3 26.7 ± 1.3 69.7 ± 2.1 86.2 ± 2.2 23.2 ± 1.1 67.7 ± 2.0 84.5 ± 2.1 33.5 ± 1.3 75.5 ± 2.1 91.3 ± 1.7 30.6 ± 1.1 71.4 ± 2.2 89.3 ± 1.8 29.8 ± 1.1 69.4 ± 2.0 87.6 ± 2.1 37.2 ± 1.4 77.3 ± 2.3 92.4 ± 1.5 26.4 ± 1.4 69.5 ± 1.9 87.3 ± 1.3 36.2 ± 1.4 77.0 ± 2.3 92.3 ± 1.8 32.5 ± 1.3 75.8 ± 2.1 90.6 ± 1.2 26.2 ± 1.3 69.5 ± 1.9 87.3 ± 1.1 31.5 ± 1.5 74.2 ± 2.3 92.4 ± 1.6 promotion by means of a two-stage carcinogenesis test of mouse skin papilloma using dimethylbenz[a]anthracene (DMBA) as initiator and TPA as promoter. Results of the test on Raji cells are reported in Table 1. Although not reported in Table 1, all compounds showed a 100% inhibitory effect at the concentration value of 1000, expressed as mol ratio/32 pmol TPA. Among the tested aldehydes, boropinal C (5) exhibited the most significant inhibitory activity on EBV-EA activation. Among the cinnamyl alcohols, boropinol A (9), boropinol C (11) and compound (16) showed similar activities to that of boropinal C (5). It’s interesting to note that secondary metabolites lacking a prenyloxy side chain like compounds 7, 8, 10, 12 and 13 are less effective as inhibitors of EBVEA activation. This suggests that the presence of a prenyloxy side chain in position 4’ on 3-phenyl-2propenal and –propenol cores enhance the inhibitory effects. The same observation was made for the activity of several other prenyloxyphenylpropanoids [4]. Results of the in vivo two-stage carcinogenesis test of mouse skin papillomas induced by DMBA and promoted by TPA using compound 15 in comparison with a positive control that developed papillomas after only 10 weeks of promotion, showed that when applied before TPA treatment, 15 delayed the formation of papillomas. In each group of animals treated with compound 16 only about 20% of mice bore papillomas at 10 weeks after promotion and even after 20 weeks of promotion 80% of the mice bore tumors. Moreover, 16 reduced the incidence of papillomas (average number of tumors per mouse): less than 5 papillomas were formed per mouse after 11 weeks of promotion and only about 3.8 papillomas 6 15 EBV-EA positive cells (% vialibility) Compd. concentration (mol ratio/32 pmol TPA) 500 100 10 27.2 ± 1.8 65.5 ± 1.0 88.5 ± 0.4 23.1 ± 1.1 62.2 ± 1.5 84.0 ± 0.3 Table 3: Inhibition of 5-LOX mediated PUFAs peroxidation by boropinic acid (15). Compds. 15 Ascorbic acid BHT Trolox a IC50 (μmol/mL)a 2.89 x 10-5 ± 2.62 x 10-6 0.105 ± 0.0072 0.023 ± 0.0052 0.047 ± 0.0048 p<0.05 at Student’s t test per mouse even after 20 weeks of promotion. The same in vitro test on EBV-EA activation was later carried out on 4’-hydroxy-3’-prenylcinnamaldehyde (6) and boropinic acid (15), which had been isolated in a second step of the ongoing studies of Itoigawa and coworkers [16]. Results are reported in Table 2. Both compounds showed a 100% inhibitory activity at a concentration value of 1000, expressed as mol ratio/32 pmol TPA, as had the previous compounds, and a good inhibitory activity on TPA-induced EBVEA activation, also at lower doses. All the other secondary metabolites that had been extracted from the roots of B. pinnata were not active in the same test. In the frame of an ongoing study devoted to the synthesis and characterization of pharmacological properties of prenyloxyphenylpropanoids, Curini and coworkers first studied the in vitro anti-inflammatory and anti-bacterial properties of boropinic acid (15). These authors found first that compound 15 did not exhibit significant antioxidant properties when submitted to the DPPH radical scavenging assay. To enforce this finding they also performed the assay for inhibition of polyunsaturated fatty acids (PUFAs) peroxidation mediated by soybean 5-lipoxygenase (5LOX), using ascorbic acid, butyl hydroxytoluene (BHT) and Trolox as positive controls [18]. Results of the latter test are reported in Table 3. Contrasting results obtained by means of chemical and enzymatic assays suggested that boropinic acid (15) acted as an effective 5-LOX inhibitor. It is noteworthy that other prenyloxyphenylpropanoids, like cinnamic acid bearing longer O-chains or coumarins, were not active in both tests. Phytochemical and pharmacological aspects of Boronia pinnata Table 4: MIC values for inhibition of growth against H. pylori by boropinic acid (15). Compd 15 Metronidazole Amoxicillin Tetracycline Clarithromycin a MIC (μg/mL)a 1.62 > 200 0.781 4.00 1.25 Values are means of three experiments. To rationalize tentatively the inhibitory mechanism observed for boropinic acid and the lack of efficacy of some other prenyloxyphenylpropanoids, Curini and coworkers inferred a possible 5-LOX/ligand docking by comparative modelling. As a result of this analysis, a peculiar feature of the modelled 5 LOX/boropinic acid complex is the possibility for the hydrophobic side chain represented by the isopentenyloxy moiety to be oriented and to enter in van der Waal’s contact with a cluster of hydrophobic amino acids. This interaction is enforced by polar interactions at the same site of the carboxylic group with Ile 857 and the amide side chain Gln 514 of the enzyme. Since these additional interactions might contribute to the enhancement of the complex stability, it may be hypothesized that the loss of activity of 5-LOX in the presence of 15 could be the result of enzyme inhibition as a consequence of stable ligand docking in the active site. The same research group studied the anti-bacterial properties of boropinic acid (15). After having screened several Gram positive and Gram negative bacterial strains, they found that 15 is an effective inhibitor in vitro of the growth of Helicobacter pylori [19]. Results of the test, performed by the agar dilution method with metronidazole, amoxicillin, tetracycline and clarithromycin as reference drugs, are reported in Table 4, expressed as minimum inhibitory concentration (MIC). Although from the data reported in Table 4 it is evident that the strain of H. pylori (namely DSMZ 4867 obtained from human gastric samples) used to perform the test is clearly resistant to metronidazole, Natural Product Communications Vol. 3 (12) 2008 2149 the activity of boropinic acid (15) as an inhibitory agent of the growth of H. pylori is comparable to that of most common antibiotics currently used in therapy to eradicate bacterial infections. Based on these preliminary results, boropinic acid (15) could be viewed as a potential lead compound for a novel class of H. pylori inhibitors. However, studies aimed to clearly depict the mechanism of action of this secondary metabolite, and in vivo tests using a suitable animal model, and in vitro and in vivo tests to evaluate the activity of 15 against strains of H. pylori isolated from clinical patients have to be carried out in the near future. Conclusions and future perspectives In this review we have reported what is known so far in the literature about the Australian shrub B. pinnata. Twenty-four secondary metabolites have been isolated in low concentrations and structurally characterized from different parts of this plant and the major part of these natural compounds belong to the class of prenyloxyphenylpropanoids. With the aim of obtaining these compounds in sufficient quantities to determine a detailed pharmacological profile, bypassing difficulties linked to the low quantities obtainable from natural sources, a few of the compounds have been obtained by chemical synthesis by means of environmentally sound, friendly and high yielding methodologies. In particular, boropinic acid has been obtained in nearly quantitative yield. Preliminary pharmacological studies on simple and oxyprenylated phenylpropanoids from B. pinnata have shown that compounds like boropinal C and boropinic acid show valuable biological properties, such as anti-cancer, anti-inflammatory and anti-ulcer activities. In the search for novel therapeutic remedies from nature, the data reported in this review will certainly prompt further studies on this plant to better define the profile of its secondary metabolites and their pharmacological properties, in particular by means of in vivo studies employing suitable animal models. References [1] Kuzuyama T, Noel JP, Richard SB, (2005) Structural basis for the promiscuous biosynthetic prenylation of aromatic natural products. Nature, 435, 983–987. [2] Haagen-Smit J. (1953) The biogenesis of terpenes. Annual Review of Plant Physiology, 4, 305–324. [3] Lichtenthaler HK. (1999) The 1-deoxy-D-xylulose-5-phosphate pathway of isoprenoid biosynthesis in plant. Annual Review of Plant Physiology. Molecular Biology, 50, 47–65. [4] Epifano F, Genovese S, Menghini L, Curini M. (2007) Chemistry and pharmacology of oxyprenylated secondary plant metabolites. Phytochemistry, 68, 939-953. 2150 Natural Product Communications Vol. 3 (12) 2008 Curini et al. [5] Nazrul Islam SK, Grey AI, Waterman PG, Ahasan M. (2002) Screening of eight alkaloids and ten flavonoids isolated from four species of the genus Boronia (Rutaceae) for antimicrobial activities against seventeen clinical microbial strains. Phytotherapy Research, 16, 672-674. [6] Ahsan M, Gray AI, Leach G, Waterman PG. (1993) Quinolone and acridone alkaloids from Boronia lanceolata. Phytochemistry, 33, 1507-1510. [7] Ahsan M, Gray AI, Waterman PG. (1994) 4-Quinolinone, 2-quinolinone, 9-acridanone and furoquinoline alkaloids from the aerial parts of Boronia bowmanii. Journal of Natural Products, 57, 670-672. [8] Ahsan M, Armstrong JA, Gibbons S, Gray AI, Waterman PG. (1994) Novel O-prenylated flavonoids from two varieties of Boronia caerulescens. Phytochemistry, 37, 259-266. [9] Ahsan M, Gray AI, Waterman PG. (1994) Farnesyl acetophenone and flavanone compounds from the aerial parts of Boronia ramose. Journal of Natural Products, 57, 673-676. [10] Ghisalberti EL. (1997) Phytochemistry of the Australian Rutaceae. Phytochemistry, 47, 163-176. [11] Bodkin F. (1993) The Essential Reference Guide to Native and Exotic Plants in Australia. In Encyclopaedia Botanica, Cornstalk Publishing, Sydney, Australia, 155-160. [12] Smith HG. (1919) Essential oil of Boronia pinnata Sm. and the presence of elemicin. Proceeding Royal Society of Victoria, 32, 14-19. [13] Welch MB, Penfold AR. (1922) Two pinnate leaf Boronias and their essential oils. Journal and Proceedings of the Royal Society of New South Wales, 56, 196-209. [14] Morrison FR. (1921) The occurrence of rutin in the leaves of the Boronia (Rutaceae). Journal and Proceedings of the Royal Society of New South Wales, 55, 210-214. [15] Ito C, Itoigawa M, Furukawa H, Ichiishi E, Mukainaka T, Okuda M, Ogata M, Tokuda H, Nishino H. (1999) Anti-tumor-promoting effects of phenylpropanoids on Epstein-Barr virus activation and two-stage mouse skin carcinogenesis. Cancer Letters, 142, 49-54. [16] Ito C, Itoigawa M, Otsuka T, Takuda H, Nishino H, Furukawa H. (2000) Constituents of Boronia pinnata. Journal of Natural Products, 63, 1344-1348. [17] Wang Q, He K, Hou Z. (2003) First synthesis of three naturally occurring O-prenylated phenylpropanoids. Youji Huaxue, 23, 182-186. [18] Curini M, Epifano F, Genovese S, Menghini L, Ricci D, Fraternale D, Giamperi L, Bucchini A. Bellacchio E. (2006) Lipoxygenase inhibitory activity of boropinic acid, active principle from Boronia pinnata. Natural Product Communications, 1, 1141-1145. [19] Epifano F, Menghini L, Pagiotti R, Angelini P, Genovese S, Curini M. (2006) In vitro inhibitory activity of boropinic acid against Helicobacter pylori. Bioorganic and Medicinal Chemistry Letters, 16, 5523-5525. NPC Natural Product Communications Therapeutic Potential of Kalanchoe Species: Flavonoids and other Secondary Metabolites 2008 Vol. 3 No. 12 2151 - 2164 Sônia S. Costaa,*, Michelle F. Muzitanoa,b, Luiza M. M. Camargoa and Marcela A. S. Coutinhoa a Núcleo de Pesquisas de Produtos Naturais, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil b Laboratório de Biologia do Reconhecer, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, RJ, Brazil sscosta@nppn.ufrj.br Received: July 22nd, 2008; Accepted: November 5th, 2008 The Kalanchoe genus (syn. Bryophyllum), family Crassulaceae, comprises 125 species, most of them native to Madagascar. The great importance of several of these species for the traditional medicine in several regions of the World, esspecially India, Africa, China and Brazil, stimulated research programs into these plants from both a pharmacological and chemical point of view. The present review focuses on the main results obtained during the last decade on the secondary metabolites isolated from these species – endowed or not with a specific biological profile – with emphasis on flavonoids. The distribution of these molecules in the genus will be summarized and special attention will be given to K. brasiliensis and K. pinnata, two species well-known for healing inflammatory and infectious processes. Ornamental Kalanchoe species are also discussed as a potential source of bioactive compounds. This review covers the period 1970-2008. Keywords: Kalanchoe, Crassulaceae, flavonoids, biological activity, Kalanchoe pinnata, Kalanchoe brasiliensis. 1. Introduction The genera Kalanchoe, Rhodiola and Sedum are three medicinally important genera in the family Crassulaceae. The genus Kalanchoe (syn. Bryophyllum), first established by Adanson (1763), comprises 125 species, most of them native to Madagascar [1]. The great importance of some Kalanchoe species for traditional medicinal use in many regions of the World, especially India, Africa, China and Brazil, stimulated several research groups to investigate the pharmacological properties of the active plant extracts as well as their chemical composition [2-17]. Several Kalanchoe species occur in Brazil, some of which, such as K. pinnata and K. brasiliensis, are used for the treatment of rheumatism, inflammation, burns, wounds and abscesses [2]. Other species such as K. fedtschenkoi and K. blossfeldiana are cultivated for ornamental purposes [18]. Indeed, a significant number of Kalanchoe species – regardless of their medicinal interest – have a great ornamental value as they exhibit extremely beautiful flowers with a broad spectrum of colors varying from white, light yellow to orange, pink, red and purple. Although Kalanchoe plants have important medicinal applications, some of them are known to be toxic to cattle [19] and chickens [20]. Flowering plants were found to be responsible for the most important forms of poisonings in Australia [19]. As far as we know, Kalanchoe flowers are not used for medicinal purposes; only leaves or non-flowering aerial parts are usually used. Succulent plants, especially those from the genus Kalanchoe, have been thoroughly studied with respect to their Crassulacean-acid metabolism (CAM) ranging from ecological, physiological and cell biological aspects, transport and compartmentalization to molecular biology [21-24]. The early studies on the secondary metabolites from Kalanchoe species mainly focused on their flavonoid content. Gaind & Gupta, at the beginning of the 1970s, accomplished the pioneer studies on the 2152 Natural Product Communications Vol. 3 (12) 2008 isolation and identification of glycosyl flavonoids from K. pinnata [25,26]. Flavonoids can occur naturally as aglycones or glycosides [27]. Plants containing flavonoids have been used for thousands of years in traditional medicine [28]. Bufadienolides - perhydrophenanthrene derivatives substituted at C-17 with a pentadienolide – are another metabolite class present in some Kalanchoe species. These compounds originate from mevalonate-isopentenyl pyrophosphate– pregnenolone metabolism [29] and are involved in poisoning of pets and cattle after ingestion of the flowers of some Kalanchoe species [20,30]. A brief review of the different classes of secondary metabolites isolated from Kalanchoe species, such as bufadienolides, terpenoids and flavonoids, as well as some biological aspects, has already been published by one of us [31]. The increasing interest in Kalanchoe species for the discovery of new bioactive substances, led us to review the state of the art in the study of their bioactive chemical composition. The present review will particularly focus on flavonoid metabolites, the main bioactive chemical constituents of plants of this genus. 2. Chemical, ethnomedicinal and potential therapeutic aspects of the genus Kalanchoe: state of the art. Among the most widespread Kalanchoe species, two of them - K. pinnata and K. brasiliensis - are widely employed in Brazil to treat different infectious, injurious and inflammatory processes [2]. These medicinal herbs are locally known under the same common names, "saiao" or "coirama", in the different Brazilian regions. In most cases, one of these species is employed in substitution for the other, generally in an undifferentiated manner. K. brasiliensis Camb. is the only native Kalanchoe species in Brazil and was the first one to be studied in depth by our group. Although the leaves are largely used in Brazil, their chemical composition and pharmacological effects were yet unknown when we started our studies. First results from our group demonstrated that K. brasiliensis (Kb) juice, aqueous and alcoholic extracts exerted a direct in vitro inhibitory effect on human lymphocyte proliferation [32]. Bioguided purification of the juice of the combined aerial parts, Costa et al. based on its in vitro lymphocyte anti-proliferative activity, led to the isolation and identification of seven patuletin rhamnosides [5]. Among them, patuletin 3-O-(4"-O-acetyl-α-L-rhamnopyranosyl)-7O-(2"'-O-acetyl-α-L-rhamnopyranoside), patuletin 3-O-α-L-rhamnopyranosyl-7-O-(2"'-O-acetyl-α-Lrhamnopyranoside) and patuletin 3-O-(4"-O-acetylα- L -rhamnopyranosyl)-7-O-rhamnopyranoside named kalambroside A, B and C, respectively, were described for the first time. The four other flavonoids were the known patuletin rhamnosides initially isolated from K. gracilis, a medicinal plant used in Taiwan for the treatment of tissue inflammation [33]. One characteristic of the main patuletin rhamnosides isolated from K. brasiliensis is the presence of either a mono or a di-acetylated sugar residue, such as those found in kalambroside A, B and C (Figure 1). The known flavonoids quercitrin and isoquercitrin were also isolated from the same specimens [34]. OH 3' OH 4' O 7 O 2 1' Me O 1''' HO 3 O 2''' MeO 6 HO OR 2 OH O Me O 1" R1 O R 1 R2 4" HO OH Kalambroside A Ac Ac Kalambroside B H Ac Kalambroside C Ac H Figure 1 - Kalambrosides A-C from K. brasiliensis From a biological point of view, among the seven flavonoids isolated, kalambroside A, B and C were shown to be potent inhibitors of lymphocyte proliferation. Diacetylated flavonoids with two rhamnosyl units, such as kalambroside A, were more active than their monoacetylated congeners kalambrosides B and C. It was generally observed that the pure components showed higher activity then either the crude flavonoid fraction or extract. Their lymphoproliferative inhibitory activities seem to be dependent on the presence of at least one acetyl group and on its position on the rhamnosyl unit of these flavonoids. Non-acetylated derivatives, such as patuletin 3-O-rhamnoside and patuletin 3,7-di-Orhamnoside, did not show any activity [5]. An hydroalcoholic extract of leaves of K. brasiliensis showed acetylcholine esterase inhibitory effects. This activity was attributed to a flavonoid mixture, containing 8-methoxyquercetin-3,7-di-Orhamnopyranoside and 8-methoxykaempferol-3,7-diO-rhamnopyranoside [35]. The authors also reported a larvicidal effect for the plant extract. Secondary metabolites from Kalanchoe species Natural Product Communications Vol. 3 (12) 2008 2153 Antimicrobial activity was described for K. brasiliensis extracts [36]. These findings support, at least partially, the popular use of the plant for healing infected wounds. any inhibitory activity on human lymphocyte proliferation [40]. The observed inhibitory activity of lymphocyte proliferation was attributed to a fraction composed of palmitic and stearic acids [11]. This activity can be correlated with an anti-inflammatory effect and could partially explain the use of the plant against inflammatory conditions. In continuation of our phytochemical investigation of the bioactive flavonoid fraction from K. pinnata leaf extract, we isolated five flavonoids, including the already reported quercitrin and afzelin. The three others were the minor methoxylated flavones 5,7,4’-trihydroxy8,3’-dimethoxyflavone, galangustin and jaceosidin, which had not previously been reported for the Kalanchoe genus [40]. These flavones are common in the Asteraceae family [41], but methoxylation at the C-8 position is rare when compared with methoxylation at C-6 or C-7 of the A ring. Despite the unusual methoxylation at C-8, there is one example described for the Crassulaceae family. This compound, a limocitrin glycoside, was isolated from Sedum acre [42]. Both of the flavonols isolated, quercitrin and afzelin, significantly inhibit human lymphocyte proliferation (IC50 = 1.0 – 10.0 μg/mL) in vitro. Leaves of K. brasiliensis are widely used in Brazil against the inflammation and allergic reactions provoked by insect bites. The anti-histaminic property of the plant extract could be observed [37]. Additionally, the antiophidic and thyroid peroxidase inhibitory activities were demonstrated [38,39]. The compounds responsible for these pharmacological activities are not yet identified [36-39]. K. pinnata (Lamarck) Persoon (=Bryophyllum pinnatum, B. calycinum, K. calycina), one of the oldest Kalanchoe species introduced to European botanical gardens, is considered native to Madagascar and has become naturalized in the tropics throughout the World. This species is certainly the most widespread member of the Kalanchoe genus, particularly in the tropical regions, and is reputed for its medicinal properties [18]. For these reasons it has been the object of many pharmacological and chemical studies. The first of these led to the characterization of some organic acids, alkanes, terpenes, sterols and waxes [31]. The occurrence in K. pinnata of the flavonoids quercetin 3-O-diarabinoside and rutin was first reported by Gaind and Gupta [25]. Although employed for the same medicinal purposes as K. brasiliensis, K. pinnata shows a very different chemical profile concerning its phenolic metabolites, as well as different biological activities. In Brazil, this plant is known, among other names, as "saiao", "folha-da-fortuna" and "folha-do-pirarucu" [18]. The main common biological activity shared both by K. pinnata and K. brasiliensis is their antiinflammatory effect and the correlation of this with the immune system. The immunomodulatory activity observed for the different types of K. pinnata leaf extracts (juice, aqueous and ethanolic extracts) led us to carry out the first biomonitored chemical investigation of this plant. The study of a hydrosoluble fraction of a K. pinnata ethanolic extract led to the isolation of its major flavonoid component, the known quercetin 3-O-α-Larabinopyranosyl (1→2)-α-L-rhamnopyranoside [40]. Antiallergic activity was reported for this quercetin diglycoside that had been previously isolated in 1986 [4]. This flavonoid, despite its high concentration in the extract, did not show The immunomodulatory ability demonstrated by K. pinnata extracts, besides its ethnomedicinal use for healing wounds and skin diseases, led us to investigate the effect of this plant on Leishmania amazonensis-infected macrophages. L. amazonensis is one of several Leishmania protozoa that cause serious endemic diseases, leishmaniases, in tropical and subtropical regions of the World. Leishmaniases may range from single cutaneous to fatal kala-azar, affecting more than 12 million people in 88 countries [43]. There is no vaccine ready for use in humans and chemotherapy still relies on toxic i.m. injections with pentavalent antimonials, Pentostam and Glucantime, or Amphotericin B [44]. Oral treatment of L. amazonensis-infected mice with aqueous leaf extract of K. pinnata (Kp) significantly reduced the lesion size and parasite load to levels comparable to i.p. injection with Glucantime [6,9]. Furthermore, an important remission of human cutaneous leishmaniasis upon oral Kp treatment, without toxic effects, has been reported in one case study [45]. A biomonitored fractionation of Kp extract has demonstrated quercitrin to be a potent antileishmanial compound, with a low toxicity profile [46]. Along 2154 Natural Product Communications Vol. 3 (12) 2008 with quercitrin, other flavonoids were isolated, such as the new kaempferol 3-O-α-L-arabinopyranosyl (1→2)-α-L-rhamnopyranoside (kapinnatoside), quercetin 3-O-α-L-arabinopyranosyl (1→2)-α-Lrhamnopyranoside and 4’,5-dihydroxy-3’,8dimethoxyflavone 7-O-β-D-glucopyranoside, all of which showed antileishmanial activity to different extents [47]. All these results show an interesting medicinal potential for the use of Kp extract and its flavonoids against leishmania. Recently, the protective effect of K. pinnata aqueous extract against fatal anaphylactic shock in mice was reported [48]. Oral protection was accompanied by a reduced production of specific IgE antibodies, reduced eosinophilia, and impaired production of the IL-5, IL-10 and TNF-α cytokines. Kp extract prevented antigen-induced mast cell degranulation and histamine release in vitro. This activity was mainly attributed to the flavonoid quercitrin previously reported from Kp extract [48]. Indeed, the phytotherapeutic potential of this species is high, as can be deduced from the wide spectrum of biological activities so far reported for it (Table 1) [6,9,11,15,16,46-59]. It is remarkable that despite the large number of reported pharmacological properties, to date, only few bioactive molecules have been identified from this species. In the perspective of a commercial use of Kp as a standardized qualitycontrolled phytotherapeutic drug there is a requirement for the definition of a specific chemical marker for this species. Quercetin 3-O-α-Larabinopyranosyl (1→2)-α-L-rhamnopyranoside, which shows a restricted occurrence in nature, could be considered as a convenient chemical marker for K. pinnata. This flavonoid is an uncommon molecule not reported to date in other plant species, except for Alphitonia philippinensis (Rhamnaceae) [60] and K. blossfeldiana flowers, as a minor metabolite [61]. Although K. pinnata is largely used as a medicinal plant and is therefore considered as safe, some toxic bufadienolides were isolated from its leaves and also from the whole plant [62-64]. A bufadienolide from K. pinnata – bryophyllin A – was shown to be a potent anti-tumor promoter inhibitor when compared with other bufadienolides isolated from K. pinnata and K. daigremontiana x tubiflora [12]. However, these substances were not detected in the K. pinnata specimens employed in our studies. Other less studied Kalanchoe species will be discussed below under chemical and pharmacological aspects. Costa et al. Table 1: Pharmacological activities and bioactive compounds reported for extracts from Kalanchoe pinnata. Pharmacological Activity Antibacterial Antidiabetic Bioactive compounds - References [49] [50] Antihypertensive - [51] Anti-inflammatory - [52-54] Antimalarial Quercitrin, quercetin 3-O-αL-arabinopyranosyl (1→ 2)α-L-rhamnopyranoside, kaempferol 3-O-α-Larabinopyranosyl (1→ 2)-αL-rhamnopyranoside and 4’,5-dihydroxy-3’, 8dimethoxyflavone 7-O- β-Dglucopyranoside - Antinociceptive - [50] Antiulcerogenic - [53] CNS depressant - [56] Hepatoprotective - [16] Immunosuppression Fatty acids [11] Muscle relaxing - [57] Neurosedative - [57] Tocolytic - [58,59] Anti-anaphylactic Quercitrin Antileishmanial [6,9,15,46,47] [55] [48] Kalanchoe gastonis-bonnieri R. Hamet & H. Perrier (= Bryophyllum gastonis-bonnieri), known popularly as “life-plant”, “donkeys-ear” and “mala-madre”, possesses large leaves that can measure up to 50 cm in length [18]. Its leaf juice is employed vaginally as a contraceptive in Mexican traditional medicine [14]. The plant is also employed against genito-urinary troubles and vaginal infections [65]. In Madagascar it is employed to treat wounds caused by insect bites [18]. The chemical composition of the juice obtained from K. gastonis-bonnieri leaves is under investigation in our laboratories. Kalanchoe gracilis Hance is used in Taiwan for the treatment of tissue inflammation [33]. Nineteen flavonoids were isolated from the aerial parts of this species, most of them exhibiting a patuletin skeleton bearing a mono or a di-acetylated sugar residue [66]. Four eupafolin rhamnosides, some of them bearing an acetyl substitution in the sugar moiety, were also reported. Luteolin and four other flavonoids (eupafolin, kaempferol, quercetin and quercitrin) were also identified [33]. Table 2 summarizes the flavonoids reported for the Kalanchoe species. Recently, new cytotoxic bufadienolides were described from K. gracilis (kalanchosides A-C) (Figure 2) [67] and K. hybrida (kalanhybrins A-C) (Figure 3) [68]. Secondary metabolites from Kalanchoe species Natural Product Communications Vol. 3 (12) 2008 2155 Table 2: Flavonoids isolated from Kalanchoe species organized by their classes and frequency in the genus. Flavonoids Glycosides Flavonols Patuletin 3,7-di-O-α-L-rhamnopyranoside Patuletin 3-O-α-L-rhamnopyranoside Kaempferol 3-O-α-L-rhamnopyranoside (afzelin) Kaempferol 3-glucoside (astragalin) Kaempferol 3-O-β-D-xylopyranosyl (1→2)-α-L-rhamnopyranoside-7-O-α-L-rhamnopyranoside (sagittatin A) Kaempferol 3-O-β-D-glucopyranoside-7-O-α-L-rhamnopyranoside Kaempferol 3-O-β-D-xylopyranosyl (1→2)-O-α-L-rhamnopyranoside Kaempferol 3-O-α-L-arabinopyranosyl (1→2)-α-L-rhamnopyranoside (kapinnatoside) Kaempferol arabinosyl-coumaroyl (bryophylloside) Quercetin 3-O-di-arabinoside Quercetin 3-O- β -D-glucopyranoside (isoquercitrin) Quercetin 3-O-α-L-arabinopyranosyl (1→2)-α-L-rhamnopyranoside Quercetin 3-O-glucoside-7-O-rhamnoside Quercetin 3-O-α-L-rhamnopyranoside (quercitrin) Quercetin 3-O-α-L-rhamnopyranosyl (1→6)-β-D-glucopyranoside (rutin) Quercetin 3-O-β-D-glucopyranosyl (1→2)-β-D-xylopyranoside Flavones Eupafolin 4’-O-rhamnoside Eupafolin 3,7-di-O-rhamnoside 4’,5-dihydroxy-3’,8-dimethoxyflavone-7-O-β-D-glucopyranoside Anthocyans Cyanidin 3-O-β-D-glucoside Cyanidin 3,5-O-β-D-diglucoside Pelargonidin 3,5-O-β-D-diglucoside Peonidin 3,5-O-β-D-diglucoside Delfinidin 3,5-O-β-D-diglucoside Petunidin 3,5-O-β-D-diglucoside Malvidin 3,5-O-β-D-diglucoside Acyl-glycosides Flavonols Patuletin 3-O-α-L-rhamnopyranosyl-7-O-(3"'-O-acetyl-α-L-rhamnopyranoside) Patuletin 3-O-α-L-rhamnopyranosyl-7-O-(4"'-O-acetyl-α-L-rhamnopyranoside) Patuletin 3-O-α-L-rhamnopyranosyl-7-O-(3"', 4"'-O-diacetyl-α-L-rhamnopyranoside Patuletin 3-O-(4"-O-acetyl-α-L-rhamnopyranosyl)- 7-O-(3"'-O-acetyl-α-L-rhamnopyranoside) Patuletin 3-O-(3"-O-acetyl-α-L-rhamnopyranosyl)- 7-O-(3"'-O-acetyl-α-L-rhamnopyranoside) Patuletin 3-O-(4"-O-acetyl-α-L-rhamnopyranosyl)-7-O-(2"'-O-acetyl-α-L-rhamnopyranoside) (kalambroside A) Patuletin 3-O-α-L-rhamnopyranosyl-7-O-(2"'-O-acetyl-α-L-rhamnopyranoside) (kalambroside B) Patuletin 3-O-(4"-O-acetyl-α-L-rhamnopyranosyl)-7-O-rhamnopyranoside (kalambroside C) Patuletin 3-O-(4"-O-acetyl-α-L-rhamnopyranosyl)-7-O-(2"', 4"'-O-diacetyl-α-L-rhamnopyranoside) Kaempferol 3-O-β-D-xylopyranosyl (1→2)-α-L-rhamnopyranoside-7-O-4""-O-acetyl-α-L-rhamnopyranoside (4””acetylsagittatin A) Flavones Eupafolin 3-O-rhamnopyranosyl-7-O-(4"'-O-acetyl-α-L-rhamnopyranoside) Eupafolin 3-O-(3"-O-acetyl-α-L-rhamnopyranosyl)-7-O-(3"'-O-acetyl-α-L-rhamnopyranoside) Aglycones Flavonols Patuletin Species Reference K. brasiliensis K. gracilis K. spathulata K. brasiliensis K. gracilis K. gracilis K. pinnata K. spathulata K. pinnata K. fedtschenkoi K. streptantha K. blossfeldiana K. fedtschenkoi K. blossfeldiana K. fedtschenkoi K. blossfeldiana K. pinnata K. daigremontiana K. pinnata K. blossfeldiana K. brasiliensis K. blossfeldiana K. pinnata K. blossfeldiana K. spathulata K. brasiliensis K. gracilis K. pinnata K. pinnata K. prolifera [5] [33] [69] [5] [33] [66] [7] [69] [25] [70] [71] [61] [70] [61] [70] [61] [47] [26] [25] [61] [5] [61] [4] [61] [69] [34] [66] [46] [25] [72] K. gracilis K. gracilis K. pinnata [66] [66] [47] K. blossfeldiana K. blossfeldiana K. blossfeldiana K. blossfeldiana K. blossfeldiana K. blossfeldiana K. blossfeldiana [26] [26], [61] [26], [61] [61] [61] [61] [61] K. brasiliensis K. gracilis K. gracilis K. gracilis K. brasiliensis K. gracilis K. gracilis K. brasiliensis K. brasiliensis K. brasiliensis K. gracilis K. streptantha [5] [33] [33] [33] [5] [33] [33] [5] [5] [5] [66] [71] K. gracilis K. gracilis [33] [33] K. gracilis K. spathulata [33] [69] 2156 Natural Product Communications Vol. 3 (12) 2008 Kaempferol K. gracilis K. pinnata K. spathulata K. crenata K. gracilis K. pinnata K. spathulata K. crenata Quercetin Flavones Eupafolin Luteolin 5,7-dihydroxy-8,4’-dimethoxy-flavone (Galangustin) 5,7,4’-trihydroxy-6,3’-dimethoxy-flavone (Jaceosidin) 5,7,4’-trihydroxy-8,3’-dimethoxy-flavone Others Flavan-3-ol Leucocyanidin R2 OHC R1 O Bufadienolides O Kalanchoside A Me Me Kalanchoside B Me OH OH OH Kalanchoside C Me H2 OH O HO OH H2 OH O OH O OH Figure 2: Bufadienolides from K. gracilis. OHC MeO Me R1O R3 H H R2O OH O OH Table 2 (Contd.) [66] [26] [69] [26] [66] [26] [69] [26] K. gracilis K. gracilis K. pinnata K. pinnata K. pinnata [66] [66] [40] [40] [40] K. pinnata K. blossfeldiana [73] [26] R2 O HO H H R 1O Costa et al. Bufadienolides R1 R2 R3 Kalanhybrin A H Ac CHO Kalanhybrin B Ac H CHO Kalanhybrin C H Ac Me Figure 3: Bufadienolides from K. hybrida. The study of a methanolic extract of the leaves of Kalanchoe streptantha Baker, one of the fifty nine Kalanchoe species reported in Madagascar, led to the new kaempferol 3-O-β-D-xylopyranosyl (1→2)-α-L-rhamnopyranoside-7-O-4""-O-acetyl-αL-rhamnopyranoside (4""-acetylsagittatin A) and sagittatin A [71]. The flavonoids 4""-acetylsagittatin A and sagittatin A, at 25 μg/mL, inhibited 50% of human lymphocyte proliferation stimulated by phytohemaglutinin A [71]. These two flavonoids showed weaker antiproliferative activity than patuletin 3-O-(4"-Oacetyl-α-L-rhamnopyranosyl)-7-O-(3"'-O-acetyl-αL-rhamnopyranoside), kalambroside A and kalambroside B, previously isolated from K. brasiliensis, for which the IC50 values ranged from 0.25 to 1.0 μg/mL [5]. In Nepal, leaves from Kalanchoe spathulata DC are used to treat burns and skin diseases [74]. Leaves and flowers from this species were reported to exhibit the same flavonoid content - patuletin 3,7-di-Orhamnoside, together with patuletin, quercetin, quercetin 3-O-glucoside-7-O-rhamnoside, kaempferol and afzelin - as reported by Gaind et al. [69]. Kalanchoe prolifera (Bowie) R. Hamet, similarly to other Kalanchoe species in Brazil, is used against rheumatism in Madagascar [18]. Razanamahefa et al. [72] isolated, from this widespread medicinal plant, quercetin-3-O-β-D-glucopyranosyl (1→2)-β-Dxylopyranoside, which was later synthesized [75]. Kalanchoe tubiflora Raym.-Hamet (= Bryophyllum tubiflorum) originates from Madagascar and has an ornamental value due to its very beautiful flowers [18]. Previous studies with K. tubiflora revealed that its leaves and flowers, when consumed by cows, caused intoxication symptoms like diarrhea [30]. The same kind of toxic event was observed for K. lanceolata (Forsskäl) Persoon in Zimbabwe [76]. Two bufadienolides were described from K. lanceolata, lanceotoxin A and B [77]. Kalanchoe crenata (Baker) R. Hamet (= K. laxiflora) is a medicinal species, whose infusions are known for their vermifugous properties [18]. In African traditional medicine it is also used as a remedy against otitis, headache, inflammations, convulsions and general debility [78]. Nguelefack et al. [17,78] suggest peripheral and central analgesic activities, as well as an anticonvulsant effect for leaf extracts of K. crenata. Aditionally, in Cameroon, this species is widely used in the treatment of diabetes mellitus. Recently, the effect of the water-ethanol extract of Secondary metabolites from Kalanchoe species Natural Product Communications Vol. 3 (12) 2008 2157 this plant on blood glucose levels was investigated in fasting normal and diet-induced diabetic rats. This study reported a significant improvement in glucose clearance and/or uptake and resistance to bodyweight gain and insulin sensitivity of K. crenata extract [79]. The bioactive compounds remain to be identified. The presence of quercetin and kaempferol in this species was early described [26]. Kalanchoe thyrsiflora Harv. has a high ornamental value and is commercialized in many parts of the world under the names of “flap-jack” or “dog-tongueplant”. Recently, in a random screening for anticancer activity of South African plant extracts, a methanolic extract from roots and leaves of K. thyrsiflora was tested against three human cell lines (breast MCF7, renal TK10 and melanoma UACC62). This extract did not show general cytotoxicity against all three cell lines, but displayed potent activity against only one of the cell lines (renal TK10). The growth inhibitory activity against the other two-cell lines was considered moderate by the authors [83]. The anticancer activity of K. thyrsiflora is supposed to be related to the bufadienolides generally occurring in Kalanchoe species. Root decoctions are traditionally used in Africa as anthelmintic enemas and administered to pregnant women who do not feel well [83]. The phenolic composition of this plant is under investigation in our laboratories, as well as four other Kalanchoe species with ornamental value. Our group is currently developing projects for the search of new molecules with pharmacological properties from flowers and leaves of some ornamental Kalanchoe species, with emphasis on flavonoids. Preliminary results are mentioned further. Kalanchoe blossfeldiana Poelln. is a dark green succulent with large umbels of flower clusters held above the foliage. This plant has a great commercial value as an ornamental. Nielsen et al. [61], in their study on the flavonoid composition of flowers from K. blossfeldiana varieties, reported the isolation of pelargonidin, cyanidin, peonidin, delphinidin, petunidin and malvidin 3,5-O-β-D-diglucosides. Pink, red and magenta varieties contain relatively high amounts of quercetin derivatives. Recently we identified the known quercetin 3,7-di-Orhamnopyranoside from the juice of K. blossfeldiana leaves, in a study searching for antiviral compounds [80]. This flavonoid was a new report for the Kalanchoe genus. Kalanchoe tomentosa Baker, a beautiful ornamental species, known as “panda plant”, is a perennial succulent covered in white felt punctuated with delightful brown patches along the leaf edges [18]. Few studies have been devoted to the chemical profile of this species [20, 81]. Another species native to Madagascar, K. daigremontiana R. Hamet & H. Perrier, is known as “mother-of-thousands” or “devil’sbackbone”. Two toxic bufadienolides with an unusual substitution pattern were reported for this species. Daigremontianin and bersaldegenin-1,3,5orthoacetate, which was also found in K. tubiflora, were shown to have pronounced sedative, positive inotropic and CNS-activities [82]. From the leaves of K. daigremontiana×tubiflora were isolated an insecticidal bufadienolide, methyl daigremonate, along with four known bufadienolides [12]. Their insecticidal activities were assessed against larvae of silkworm (Bombyx mori). Kalanchoe fedtschenkoi R. Hamet & H. Perrier is an ornamental plant widespread in Brazil and reputed to be toxic to chickens [20]. This species was exhaustively studied for its CAM metabolism [84]. The flavonoid composition of leaves infusion from K. fedtschenkoi has been studied in our laboratories. Two main fractions (F2-C and F2-E) were purified and fractionated to give three flavonoids: kaempferol-3-O-β-D-xylopyranosyl (1→2)-O-α-L-rhamnopyranoside, kaempferol-3-O-βD-glucopyranoside-7-O-α-L-rhamnopyranoside and sagittatin A. This last compound was previously isolated from K. streptantha [71]. Crude extract as well as fractions F2-C and F2-E exhibited strong cytotoxic activities, especially against resistant leukemia cells. Fraction F2-C reduced cellular viability of both parental tumor line and the resistant tumor line, besides presenting low toxicity in renal cells [70]. 3. Flavonoids and their distribution in the genus Kalanchoe Although there has been an increasing interest in the biological activities of Kalanchoe extracts in the last 20 years, little progress was made towards the chemical identification of the bioactive components. It is unquestionable that flavonoids are the most important chemical class in Kalanchoe species, 2158 Natural Product Communications Vol. 3 (12) 2008 Costa et al. Table 3: Bioactive flavonoids from Kalanchoe species. Entries were organized in the same sequence used for Table 2. Flavonoids Glycosides Some biological activities Reference Flavonols Patuletin 3-O-α-L-rhamnopyranosyl-7-O-α-L-rhamnopyranoside Lymphocyte growth inhibitor [5] Patuletin 3-O-α-L-rhamnopyranoside Kaempferol 3-O-rhamnoside (afzelin) Lymphocyte growth inhibitor Lymphocyte growth inhibitor, antimalarial, antioxidant, anti-complement activity Antiallergic Antileishmanial [5] Kaempferol 3-glucoside (astragalin) Kaempferol 3-O-α-L-arabinopyranosyl (1→2)-α-L-rhamnopyranoside Quercetin 3-O-β-D-glucopyranoside (isoquercitrin) Antioxidant, antiprotozoal Quercetin 3-O-α-L-arabinopyranosyl (1→2)-α-L-rhamnopyranoside Antiallergic, antileishmanial Quercetin 3-O-α-L-rhamnopyranoside (quercitrin) Antiviral, antileishmanial, anti-anaphylactic, anti-complement, antioxidant Antiviral, antiallergic, antioxidant, anti-inflammatory, antiulcer, gastro-protective Quercetin 3-O-β-L-rhamnopyranosyl (1→6)-β-D-glucopyranoside (rutin) [7, 85-87] [88] [47] [89, 90] [4, 47] [28, 46, 48, 87, 91] [28, 92, 93] Flavones 4’,5-dihydroxy-3’,8-dimethoxyflavone-7-O-β-D-glucopyranoside Anthocyans Antileishmanial Cyanidin 3-O-glucoside Skin photoprotective agent, antioxidant [47] [94, 95] Acyl-glycosides Flavonols Patuletin 3-O-(4"-O-acetyl-α-L-rhamnopyranosyl)- 7-O-(3"'-O-acetyl-α-Lrhamnopyranoside) Patuletin 3-O-(4"-O-acetyl-α-L-rhamnopyranosyl)- 7-O-(2"'-O-acetyl-α-Lrhamnopyranoside) Patuletin 3-O-α-L-rhamnopyranosyl-7-O-(2"'-O-acetyl-α-Lrhamnopyranoside) (kalambroside B) Patuletin 3-O-(4"-O-acetyl-α-L-rhamnopyranosyl)-7-O-rhamnopyranoside (kalambroside C) Kaempferol 3-O-β-D-xylopyranosyl (1→2)-α-L-rhamnopyranoside-7-O- 4""O-acetyl-α-L-rhamnopyranoside Aglycones Lymphocyte growth inhibitor [5] Lymphocyte growth inhibitor [5] Lymphocyte growth inhibitor [5] Lymphocyte growth inhibitor [5] Lymphocyte growth inhibitor [71] Flavonols Patuletin Kaempferol Quercetin Antiplatelet activity, antioxidant, aldose reductase inhibitor, lipoxygenase and cyclooxygenase inhibitor, antimicrobial Antiulcer, antiinflammatory, antiallergic, antinociceptive, neuroprotective, antiatherogenic, inhibition of HL-60 cell growth, induces apoptosis in human lung non-small carcinoma cells, antioxidant Antioxidant, spasmolytic effect (smooth muscle relaxation), antiviral, anti-inflammatory, antiulcer, antiallergic, hepatoprotector [96-100] [28, 93, 101-106] [28, 93, 107] Flavones Eupafolin Luteolin 5,7,4’-trihydroxy-6,3’-dimethoxy-flavone (Jaceosidin) Hepatoprotector, antiproliferative activity against HeLa cell, antioxidant, cardiopreventive Anti-inflammatory, hepatoprotector, antiallergic, vascular relaxation, antioxidant Anti-inflammatory, citotoxicity to MCF10A-ras cells, oncogene inhibitor, antiallergic [108-110] [106, 111] [112-114] Others Flavan-3-ol regarding the number of reports and biological activities. The presence of flavonols, flavones, anthocyanins, leucoanthocyanins and flavan-3-ols has been reported by several authors. Flavonols are the most frequent flavonoids in Kalanchoe species, representing aproximately 60% (Figure 4). Patuletin, kaempferol and quercetin are the most reported aglycones (Figures 5 and 6). Antioxidant, anticarcinogen, cardiopreventive, antimicrobial, antiviral, neuroprotective, anti-amyloidogenic effect [115, 116] The distribution of Kalanchoe flavonoids following their respective parent aglycone is shown in Table 4. Table 2 presents all flavonoids reported for Kalanchoe species, while Table 3 shows only those for which specific bioactivities have been reported. Several authors reported biological activities for patuletin aglycone, among them, antiplatelet activity, Secondary metabolites from Kalanchoe species 70 Flavan-3-ol (01) Leucocyanidin (01) Malvidin (01) Petunidin (01) 40 Delfinidin (01) 30 Peonidin (01) Pelargonidin (01) (29) 60 Occurrence (%) Natural Product Communications Vol. 3 (12) 2008 2159 50 (10) 20 (07) Cyanidin 10 (01) Galangustin (01) Luteolin (01) 5.7.4'-tri-OH-8.3'-di-OMe-flavone (01) Fl av on es A nt ho cy an Le s uc oc ya ni di n Fl av an -3 -o ls Fl av on ol s 0 Eupafolin OH Patuletin Quercetin Kaempferol Eupafolin R3 R2 OH O R1 R 2 R 3 OH OMe OH OH H OH H H OH OH OMe H OH OH HO O OH OH (07) Kaempferol (08) Patuletin (03) 0 10 20 30 Occurrence (%) Figure 6: The different aglycones reported for Kalanchoe flavonoids. The number of Kalanchoe species corresponding to each aglycone reported is shown in brackets. R1 O (01) Quercetin Figure 4: The distribution of flavonoid classes in the Kalanchoe genus. The number of flavonoids corresponding to each class is shown in brackets. HO (01) (01) Jaceosidin (01) Cyanidin Figure 5: The most common flavonoid aglycones reported for Kalanchoe species oxidative stress protection, aldose reductase inhibition, lipoxygenase and cyclooxygenase inhibition [96-99]. Excluding the first one, all the activities described are involved with antiinflamatory processes. Patuletin glycosides and acetyl-glycosides reported for K. brasiliensis and K. gracilis were less explored for their biological activities. Lymphocyte antiproliferative activity was reported for K. brasiliensis patuletin acetyl-glycosides. This inhibitory activity on the immune system could be useful in situations where an intensive immune response is undesirable, as in chronic inflammation, organ transplantation and auto-immune diseases. The second more reported aglycone in Kalanchoe species is kaempferol. Several studies focusing on antiulcer, antiinflammatory, bactericidal, antiallergic, antinociceptive, neuro-protective, antiatherogenic, HL-60 cell growth inhibitory, human lung non-small carcinoma cells apoptosis induction and antioxidant activities have been reported for this flavonoid Table 4: Occurrence of Kalanchoe flavonoids based on their respective parent aglycones. Parent aglycone Patuletin Kaempferol Quercetin Eupafolin Luteolin Galangustin Jaceosidin 5,7,4’-tri-OH8,3’-diOMeflavone Cyanidin Pelargonidin Peonidin Delfinidin Petunidin Malvidin Leucocyanidin Flavan-3-ol K. blo 3 3 - K. bra 7 2 - K. cre 1 1 - K. dai 1 - K. fed 3 - K. gra 9 2 2 5 1 - K. pin 4 5 1 1 2 K. pro 1 - K. spa 2 2 2 - K. str 2 - 2 1 1 1 1 1 1 - - - - - - 1 - - - ⇒ - = no reports; K. blo= K. blossfeldiana, K. bra= K. brasiliensis, K. cre= K. crenata (=K. laxiflora), K. dai= K. daigremontiana, K. fed= K. fedtschenkoi, K. gra= K. gracilis, K.pin= K. pinnata, K.pro= K. prolifera, K. spa= K. spathulata, K. str= K. strepthanta. (Table 3). Quercetin and its glycosides exert antiinflammatory, antiulcer, antiallergic, hepatoprotective, antiviral and antileishmanial effects, as can be seen in Table 3. The efficacy of quercetin as an anti-inflammatory and immunosuppressor agent has been extensively demonstrated [117,118]. Mookerjee et al. (1986) [119] reported the reversible lymphoproliferative inhibition effect of flavonoids, such as quercetin, in response to phytomitogens, soluble antigens, and phorbol esters by blocking events that follow the exposure to the stimulus. This activity profile was also observed for K. pinnata quercetin glycosides, as mentioned before. K. pinnata is the richest species with respect to the number of flavonoids possessing a quercetin skeleton. 2160 Natural Product Communications Vol. 3 (12) 2008 Our group has been contributing to a better knowledge of the therapeutic potential of Kalanchoe species in an interdisciplinary approach that correlates chemical and pharmacological studies. These works afford interesting molecules from the point of view of pharmaceutical application, as for example, the antileishmanial flavonoids from K. pinnata. Flavonoids possess several biological activities, including antimicrobial, antiviral, antiinflammatory and immunomodulatory [28]. Regarding this bioactive metabolite class, the genus Kalanchoe is particularly fruitful, leading to many interesting preliminary results that remain to be explored. In conclusion, the present review summarizes the chemical and biological data reported for Kalanchoe species during these last decades, with special attention to their flavonoid profile. The so far accumulated data show that Kalanchoe is a promising Costa et al. and rich genus, not only in terms of its outstanding ornamental value, but also in term of its numerous pharmaceutical promising perspectives. A few species from this genus have been studied in interdisciplinary approaches in view of finding new bioactive compounds. Much remains to be done for a better understanding of the ethnopharmacological use of various Kalanchoe species and to elucidate the chemical composition of the substances responsible for their pharmacological activities. We are pursuing our efforts in studying Kalanchoe species not yet explored with these perspectives in mind. Acknowledgments - We are grateful to all students and research partners for their contribution to the studies reported here. We thank Dr J-P Férézou (Ecole Polytechnique, France) for helpful suggestions and advice. Thanks to CNPq, CAPES, FAPERJ and SUS (Ministério da Saúde) for grants and fellowships. References [1] Allorge-Boiteau L. (1996) Madagascar centre de speciation et d'origine du genre Kalanchoe (Crassulaceae). In Biogéographie de Madagascar. Lourenço WR (Ed). Éditions de l'ORSTOM, Paris, 137-145. [2] Lucas V, Machado O. (1946) Contribuição ao estudo das plantas medicinais brasileiras-Saião. Revista da Flora Medicinal, 77, 1-39. [3] Lipetz J. (1967) Wound healing in Kalanchoe. Annals of the New York Academy of Sciences, 144, 320-325. [4] Ichikawa M, Ogura M, Lijima T. (1986) Antiallergic flavone glycoside from Kalanchoe pinnatum. Patent 61, 118, 396 [86, 118, 319] (Cl. CO7H17/07), Apl 84/240, 282, 14 nov 1984, 4p-Chemical Abstracts, 105, 178423q. [5] Costa SS, Jossang A, Bodo B, Souza MLM, Moraes VLG. (1994) Patuletin acetylrhamnosides from Kalanchoe brasiliensis as inhibitors of human lymphocyte proliferative activity. Journal of Natural Products, 57, 1503-1510. [6] Da Silva SA, Costa SS, Mendonça SC, Silva EM, Moraes VL, Rossi-Bergmann B. (1995) Therapeutic effect of oral Kalanchoe pinnata leaf extract in murine leishmaniasis. Acta Tropica, 60, 201-210. [7] Rossi-Bergmann B, Costa SS, Moraes VLG. (1997) Brazilian medicinal plants: a rich source of immunomodulatory substances. Ciência e Cultura, 49, 395-401. [8] Mourão RH, Santos FO, Franzotti EM, Moreno MP, Antoniolli AR. (1999) Antiinflammatory activity and acute toxicity (LD50) of the juice of Kalanchoe brasiliensis Camb. leaves picked before and during blooming. Phytotherapy Research, 13, 352-354. [9] Da Silva SA, Costa SS, Rossi-Bergmann B. (1999) The anti-leishmanial effect of Kalanchoe is mediated by nitric oxide intermediates. Parasitology, 118, 575-582. [10] Umbuzeiro-Valent G, Roubicek DA, Haebisch EM. (1999) Mutagenic and antimutagenic evaluation of the juice of the leaves of Bryophyllum calycinum (Kalanchoe pinnata), a plant with antihistamine activity. Environmental and Molecular Mutagenesis, 33, 325-327. [11] Almeida AP, Da Silva SA, Souza ML, Lima LM, Rossi-Bergmann B, Moraes VLG, Costa SS. (2000) Isolation and chemical analysis of a fatty acid fraction of Kalanchoe pinnata with a potent lymphocyte suppressive activity. Planta Medica, 66, 134-137. [12] Supratman U, Fujita T, Akiyama K, Hayashi H, Murakami A, Sakai H, Koshimizu K, Ohigashi H. (2001) Anti-tumor promoting activity of bufadienolides from Kalanchoe pinnata and K. daigremontiana x tubiflora. Bioscience, Biotechnology, and Biochemistry, 65, 947-949. [13] Ibrahim T, Cunha JMT, Madi K, Da Fonseca LMB, Costa SS, Koatz VLG. (2002) Immunomodulatory and anti-inflammatory effects of Kalanchoe brasiliensis. International Immunopharmacology, 2, 875-883. [14] Miranda-Beltrán ML, Puebla-Pérez AM, Guzmán-Sánchez A, Huacuja-Ruiz L. (2003) Male rat infertility induction/spermatozoa and epididymal plasma abnormalities after oral administration of Kalanchoe gastonis bonnieri natural juice. Phytotherapy Research, 17, 315-319. [15] Torres-Santos EC, Da Silva SA, Costa SS, Santos AP, Almeida AP, Rossi-Bergmann B. (2003) Toxicological analysis and effectiveness of oral Kalanchoe pinnata on a human case of cutaneous leishmaniasis. Phytotherapy Research, 17, 801-803. Secondary metabolites from Kalanchoe species Natural Product Communications Vol. 3 (12) 2008 2161 [16] Yadav NP, Dixit VK. (2003) Hepatoprotective activity of leaves of Kalanchoe pinnata Pers. Journal of Ethnopharmacology, 86, 197-202. [17] Nguelefack TB, Fotio AL, Watcho P, Wansi SL, Dimo T, Kamanyi A. (2004) Analgesic properties of the aqueous and ethanol extracts of the leaves of Kalanchoe crenata (Crassulaceae). Phytotherapy Research, 18, 385-388. [18] Boiteau P, Allorge-Boiteau L. (1995) Kalanchoe (Crassulacées) de Madagascar. Systématique, écophysiologie et phytochimie. Karthala, Paris. [19] McKenzie RA, Dunster PJ. (1986) Hearts and flowers: Bryophyllum poisoning of cattle. Australian Veterinary Journal, 63, 222227. [20] Williams MC, Smith MC. (1984) Toxicity of Kalanchoe spp to chicks. American Journal of Veterinary Research, 45, 543-546. [21] Brulfert J, Ravelomanana D, Kluge M. (1995) Ecophysiologie des Kalanchoe. Le métabolisme acide des Crassulacées (CAM), une voie photosynthétique adaptative. In Kalanchoe (Crassulacées) de Madagascar. Systématique, écophysiologie et phytochimie. Allorge-Boiteau L (Ed). Karthala, Paris, 207-218. [22] Winter K, Richter A, Engelbrecht B, Pousada J, Virgo A, Popp M. (1997) Effect of elevated CO2 on growth and crassulacean-acidmetabolism activity of Kalanchoe pinnata under tropical conditions. Planta, 201, 389-396. [23] Cheffings CM, Pantoja O, Ashcroft FM, Smith JAC. (1997) Malate transport and vacuolar ion channels in CAM plants. Journal of Experimental Botany, 48, 623-631. [24] Lüttge U. (2004) Ecophysiology of Crassulacean acid metabolism (CAM). Annals of Botany, 93, 629-652. [25] Gaind KN, Gupta RL. (1971) Flavonoid glycosides from Kalanchoe pinnata. Planta Medica, 20, 368-373. [26] Gaind KN, Gupta RL. (1973) Phenolic components from the leaves of Kalanchoe pinnata. Planta Medica, 23, 149-153. [27] Veitch NC, Grayer REJ. (2008) Flavonoids and their glycosides, including anthocyanins. Natural Product Reports, 25, 555-611. [28] Middleton E, Kandaswami C, Theoharides TC. (2000) The effects of plant flavonoids on mammalian cells: implications for inflammation, heart disease, and cancer. Pharmacological Reviews, 52, 673-751. [29] Steyn PS, Van Heerden FR. (1998) Bufadienolides of plant and animal origin. Natural Products Reports, 15, 397-413. [30] McKenzie RA, Franke FP, Dunster PJ. (1987) The toxicity to cattle and bufadienolide content of six Bryophyllum species. Australian Veterinary Journal, 64, 298-301. [31] Costa SS, Jossang A, Bodo B. (1995) Propriétés biologiques et phytochimie des Kalanchoe. In Kalanchoe (Crassulacées) de Madagascar. Systématique, écophysiologie et phytochimie. Allorge-Boiteau L (Ed). Karthala, Paris, 219-235. [32] Moraes VLG, Santos LFM, Castro SB, Loureiro LH, Lima OA, Lima MLM, Yien LMK, Rossi-Bergmann B, Costa SS. (1994) Inhibition of lymphocyte activation by extracts and fractions of Kalanchoe, Alternanthera, Paullinia and Mikania species. Phytomedicine, 1, 199-204. [33] Liu KCS, Lin YS, Roberts MF, Phillipson JD. (1989a) Flavonol glycosides with acetyl substitution from Kalanchoe gracilis. Phytochemistry, 28, 2813-2818. [34] Souza MLM. (1995) Estudo químico de flavonóides isolados de Kalanchoe brasiliensis Camb. com atividade sobre o sistema imunológico. MSc Thesis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil. [35] Trevisan MTS, Bezerra MZB, Santiago GMP, Feitosa CM. (2006) Atividades larvicida e anticolinesterásica de plantas do gênero Kalanchoe. Química Nova, 29, 415-418. [36] De Cunha AGM, Maia AAB, Néri EDR, Nogueira NAP, Matos FJA. (1995) Antimicrobial activity of medicinal plants. Revista Brasileira de Farmácia, 76, 5-6. [37] De Almeida CCS, Bogossian FB, Doi APK, Isoyama D, Ho Sih G, Zanolla WR, Nassis CZ. (1997) Study of antihistamine activity of Kalanchoe brasiliensis. Arquivos Brasileiros de Cardiologia, 20, 7-10. [38] Fonseca FV, Melo MM, Silva J, Pereira GP, Dantas-Barros AM. (2004) Extratos de Curcuma longa L. e Kalanchoe brasiliensis Camb. no tratamento local do envenenamento por Bothrops alternatus. Revista Brasileira de Farmacognosia, 14, 26-29. [39] Ferreira ACF, Rosenthal D, Carvalho DP. (2000) Thyroid peroxidase inhibition by Kalanchoe brasiliensis aqueous extract. Food and Chemical Toxicology, 38, 417-421. [40] Almeida AP. (2000) Extratos de Kalanchoe pinnata (Crassulaceae), uma planta usada na medicina popular: estudo químico guiado pelo ensaio de proliferação de linfócitos. PhD Thesis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil. [41] Reynaud J, Raynaud J. (1986) Les flavonoïdes de Doronicum grandiflorum. Biochemical Systematics and Ecology, 14, 191-193. [42] Stevens JF, Hart H, Elema ET, Bolck A. (1996) Flavonoid variation in eurasian Sedum and Sempervivum. Phytochemistry, 41, 503512. [43] World Health Organization (WHO), Division of http://www.who.int/tdr/diseases/leish/diseaseinfo.htm (accessed in June 2008). [44] Croft SL, Yardley V. (2002) Chemotherapy of Leishmaniasis. Current Pharmaceutical Design, 8, 319-342. [45] Torres-Santos EC, Da Silva SAG, Costa SS, Santos APPT, Almeida AP, Rossi-Bergmann B. (2003) Toxicological analysis and effectiveness of oral Kalanchoe pinnata on a human case of cutaneous Leishmaniasis. Phytotherapy Research, 17, 801-803. Control of Tropical Diseases, 2162 Natural Product Communications Vol. 3 (12) 2008 Costa et al. [46] Muzitano MF, Cruz EA, Almeida AP, Silva SAG, Kaiser CR, Guette C, Rossi-Bergmann B, Costa SS. (2006) Quercitrin: an antileishmanial flavonoid glycoside from Kalanchoe pinnata. Planta Medica, 72, 81-83. [47] Muzitano MF, Tinoco LW, Guette C, Kaiser CR, Rossi-Bergmann B, Costa SS. (2006) Assessment of antileishmanial activity of new and unusual flavonoids from Kalanchoe pinnata. Phytochemistry, 67, 2071-2077. [48] Cruz EA, Da-Silva SAG, Muzitano MF, Silva PMR, Costa SS, Rossi-Bergmann B. (2008) Immunomodulatory pretreatment with Kalanchoe pinnata extract and its quercitrin flavonoid effectively protects mice against fatal anaphylactic shock. International Immunopharmacology, 8, 1616–1621. [49] Akinpelu DA. (2000) Antimicrobial activity of Bryophyllum pinnatum leaves. Fitoterapia, 71, 193-194. [50] Ojewole JAO. (2005) Antinociceptive, anti-inflammatory and antidiabetic effects of Bryophyllum pinnatum (Crassulaceae) leaf aqueous extract. Journal of Ethnopharmacology, 99, 13-19. [51] Ojewole JAO. (2002) Antihypertensive properties of Bryophyllum pinnatum (Lam) Oken leaf extracts. American Journal of Hypertension, 15, 34A. [52] Pal S, Chaudhuri AKN. (1990) Anti-inflammatory action of Bryophyllum pinnatum leaf extract. Fitoterapia, 6, 527-533. [53] Pal S, Chaudhuri AKN. (1991) Studies on the antiulcer activity of a Bryophyllum pinnatum leaf extract in experimental animals. Journal of Ethnopharmacology, 33, 97-102. [54] Hema D, Tidjani M, Bassene E, Pousset L, Giono-Barber H. (1986) Plantes médicinales africaines XXIV. Étude de l’action antiinflammatoire de Bryophyllum pinnatum (Crassulacées). Plantes Médicinales et Phytothérapie, 3, 231-235. [55] Muñoz V, Sauvain M, Bourdy G, Callapa J, Rojas I, Vargas L, Tae A, Deharo E. (2000) The search for natural bioactive compounds through a multidisciplinary approach in Bolivia. Part II. Antimalarial activity of some plants used by Mosetene Indians. Journal of Ethnopharmacology, 69, 139-155. [56] Pal S, Sen T, Chaudhuri AKN. (1999) Neuropsychopharmacological profile of the methanolic fraction of Bryophyllum pinnatum leaf extract. Journal of Pharmacy and Pharmacology, 51, 313-318. [57] Yemitan OK, Salahdeen HM. (2005) Neurosedative and muscle relaxant activities of aqueous extract of Bryophyllum pinnatum. Fitoterapia, 76, 187-193. [58] Gwehenberger B, Rist L, Huch R, von Mandach U. (2004) Effect of Bryophyllum pinnatum versus fenoterol on uterine contractility. European Journal of Obstetrics & Gynecology and Reproductive Biology, 113, 164-171. [59] Plangger N, Rist L, Zimmermann R, von Mandach U. (2006) Intravenous tocolysis with Bryophyllum pinnatum is better tolerated than beta-agonist application. European Journal of Obstetrics & Gynecology and Reproductive Biology, 124, 168-172. [60] Jou SJ, Chen CH, Guh JH, Lee CN, Lee SS. (2004) Flavonol glycosides and cytotoxic triterpenoids from Alphitonia philippinensis. Journal of the Chinese Chemical Society, 51, 827-834. [61] Nielsen AH, Olsen CE, Moller BL. (2005) Flavonoids in flowers of 16 Kalanchoe blossfeldiana varieties. Phytochemistry, 66, 2829-2835. [62] Yamagishi T, Yan XZ, Wu RY, Mcphail DR, Mcphail AT, Lee KH. (1988) Structure and stereochemistry of bryophyllin-A, a novel potent cytotoxic bufadienolide orthoacetate from Bryophyllum pinnatum. Chemical and Pharmaceutical Bulletin, 36, 16151617. [63] Yamagishi T, Haruna M, Yan X-Z, Chang J-J, Lee K-H. (1989) Antitumor Agents, 110. 1,2 Bryophyllin B, a novel potent cytotoxic bufadienolide from Bryophyllum pinnatum. Journal of Natural Products, 52, 1071-1079. [64] Supratman U, Fujita T, Akiyama K, Hayashi H. (2000) New insecticidal bufadienolide, bryophyllin C, from Kalanchoe pinnata. Bioscience, Biotechnology, and Biochemistry, 64, 1310-1312. [65] Ososki AL, Lohr P, Reiff M, Balick MJ, Kronenberg F, Fugh-Berman A, O'Connor B. (2002) Ethnobotanical literature survey of medicinal plants in the Dominican Republic used for women's health conditions. Journal of Ethnopharmacology, 79, 285-298. [66] Liu KCS, Yang SL, Roberts MF, Phillipson JD. (1989b) Eupafolin rhamnosides from Kalanchoe gracilis. Journal of Natural Products, 52, 970-974. [67] Wu P-L, Hsu Y-L, Wu T-S, Bastow KF, Lee K-H. (2006) Kalanchosides A-C, new cytotoxic bufadienolides from the aerial parts of Kalanchoe gracilis. Organic Letters, 8, 5207-5210. [68] Kuo P-C, Kuo T-H, Su C-R, Liou M-J, Wu T-S. (2008) Cytotoxic principles and α-pyrone ring-opening derivatives of bufadienolides from Kalanchoe hybrida. Tetrahedron, 64, 3392-3396. [69] Gaind KN, Singla AK, Wallace W. (1981) Flavonoid glycosides of Kalanchoe spathulata. Phytochemistry, 20, 530-531. [70] Castricini SD. (2004) Estudo fitoquímico de Kalanchoe fedtschenkoi (Crassulaceae) e avaliação do efeito de suas frações flavonoídicas em células tumorais. MSc Thesis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil [71] Costa SS, Jossang A, Bodo B. (1996) 4””-Acetylsagittatin A, a kaempferol triglycoside from Kalanchoe streptantha. Journal of Natural Products, 59, 327-329. [72] Razanamahefa B, Demetzos C, Skaltsounis AL, Andriantisiferana M, Tillequin F. (1994) Structure and synthesis of a quercetin glucoxyloside from Kalanchoe prolifera Raym-Hamet. Heterocycles, 38, 357-373. Secondary metabolites from Kalanchoe species Natural Product Communications Vol. 3 (12) 2008 2163 [73] Ogumgbamila FO, Onawunmi GO, Andeosun O. (1997) A new acylated flavan-3-ol from Bryophyllum pinnatum. Natural Products Letters, 10, 201-203. [74] Joshi AR, Edington JM. (1990) The use of medicinal plants by two village communities in the central development region of Nepal. Economic Botany, 44, 71-83. [75] Demetzos C, Skaltsounis AL, Razanamahefa B, Tillequin F. (1994) Synthesis of quercetin-3-O-β-D-glucopyranosyl-(1→2)-β-Dxylopyranoside via orthoester methodology. Journal of Natural Products, 57, 1234-1238. [76] Anderson LA, Schultz RA, Joubert JP, Prozesky L, Kellerman TS, Erasmus GL, Procos J. (1983) Krimpsiekte and acute cardiac glycoside poisoning in sheep caused by bufadienolides from the plant Kalanchoe lanceolata Forsk. Onderstepoort Journal of Veterinary Research, 50, 295-300. [77] Anderson LAP, Steyn PS, Van Heerden FR. (1984) The characterization of two novel bufadienolides, lanceotoxin A and B from Kalanchoe lanceolata [Forssk] Pers. Journal of the Chemical Society, Perkin Transactions 1, 1573-1575. [78] Nguelefack TB, Nana P, Atsamo AD, Dimo T, Watcho P, Dongmo AB, Tapondjou LA, Njamen D, Wansi SL, Kamanyi A. (2006) Analgesic and anticonvulsant effects of extracts from the leaves of Kalanchoe crenata (Andrews) Haworth (Crassulaceae). Journal of Ethnopharmacology, 106, 70-75. Kamgang R, Mboumi RY, Fondjo AF, Tagne MA, N’dillé GP, Yonkeu JN. (2008) Antihyperglycaemic potential of the waterethanol extract of Kalanchoe crenata (Crassulaceae). Natural Medicine, 62, 34-40. [79] [80] Vargas CS. (2008) Identificación de sustancias con actividad antiviral contra el Herpes simplex tipo 1 (HSV-1) y tipo 2 (HSV-2) presentes en las flores y las hojas de Kalanchoe blossfeldiana Poelln (Crassulaceae). BSc Thesis, Pontificia Universidad Javeriana, Bogota, Colômbia. [81] Rasonaivo P, Galeffi C, Multari G, Nicoletti M, Capolongo L. (1993) Kalanchoside, a cytotoxic bufadienolidic glycoside from Kalanchoe tomentosa Baker. Gazzeta Chimica Italiana, 123, 539-541. [82] Wagner H, Fischer M, Lotter H. (1985) Isolation and structure determination of daigremontianin, a novel bufadienolide from Kalanchoe daigremontiana. Planta Medica, 169-170. [83] Fouche G, Cragg GM, Pillay G, Kolesnikova N, Maharaj VJ, Senabe J. (2008) In vitro anticancer screening of South African plants. Journal of Ethnopharmacology, 119, 455–461. [84] Geydan TD, Melgarejo LM. (2005) Crassulacean acid metabolism. Acta Biologica Colombiana, 10, 3-15. [85] Liu Y, Murakami N, Ji H, Abreu P, Zhang S. (2007) Antimalarial flavonol glycosides from Euphorbia hirta. Pharmaceutical Biology, 45, 278-281. [86] Ribeiro AB, Bolzani VD, Yoshida M, Santos LS, Eberlin MN, Silva DHS. (2005) A new neolignan and antioxidant phenols from Nectandra grandiflora. Journal of the Brazilian Chemical Society, 16, 526-530. [87] Min BS, Lee SY, Kim JH, Lee JK, Kim TJ, Kim DH, Kim YH, Joung H, Lee HK, Nakamura N, Miyashiro H, Hattori M. (2003) Anti-complement activity of constituents from the stem-bark of Juglans mandshurica. Biological & Pharmaceutical Bulletin, 26, 1042-1044 [88] Kotani M, Matsumoto M, Fujita A, Higa S, Wang W, Suemura M, Kishimoto T, Tanaka T. (2000) Persimmon leaf extract and astragalin inhibit development of dermatitis and IgE elevation in NC/Nga mice. Journal of Allergy and Clinical Immunology, 106, 159-166. [89] Liu X, Cui C, Zhao M, Wang J, Luo W, Yang B, Jiang Y. (2008) Identification of phenolics in the fruit of emblica (Phyllanthus emblica L.) and their antioxidant activities. Food Chemistry, 109, 909-915. [90] Calzada F, Alanis AD. (2007) Additional antiprotozoal flavonol glycosides of the aerial parts of Helianthemum glomeratum. Phytotherapy Research, 21, 78-80. [91] Wagner C, Fachinetto R, Dalla Corte CL, Brito VB, Severo D, de Oliveira Costa Dias G, Morel AF, Nogueira CW, Rocha JB. (2006) Quercitrin, a glycoside form of quercetin, prevents lipid peroxidation in vitro. Brain Research, 1107, 192-198. [92] Rotelli AE, Guardia T, Juárez AO, De la Rocha NE, Pelzer LE. (2003) Comparative study of flavonoids in experimental models of inflammation. Pharmacological Research, 48, 601-606. [93] Borrelli F, Izzo AA. (2000) The plant kingdom as a source of anti-ulcer remedies. Phytotherapy Research, 14, 581-591. [94] Cimino F, Ambra R, Canali R, Saija A, Virgili F. (2006) Effect of cyanidin-3-O-glucoside on UVB-induced response in human keratinocytes. Journal of Agriculture and Food Chemistry, 54, 4041-4047. [95] Serraino I, Dugo L, Mondello L, Mazzon E, Dugo G, Caputi AP, Cuzzocreo S. (2003) Protective effects of cyanidin-3-O-glucoside from blackberry extract against peroxynitrite-induced endothelial dysfunction and vascular failure. Life Sciences, 73, 1097-1114. [96] Koleckar V, Brojerova E, Rehakova Z, Kubikova K, Cervenka F, Kuca K, Jun D, Hronek M, Opletalova V, Opletal L. (2008) In vitro antiplatelet activity of flavonoids from Leuzea carthamoides. Drug and Chemical Toxicology, 31, 27-35. [97] Kim SR, Park MJ, Lee MK, Sung SH, Park EJ, Kim J, Kim SY, Oh TH, Markelonis GJ, Kim YC. (2002) Flavonoids of Inula britannica protect cultured cortical cells from necrotic cell death induced by glutamate. Free Radical Biology & Medicine, 32, 596-604. [98] Li S, Mao W, Cao X, Liang S, Ding Z, Li N. (1991) Inhibition of rat lens aldose reductase by quercetagetin and patuletin. Yan Ke Xue Bao, 7, 29-33. 2164 Natural Product Communications Vol. 3 (12) 2008 Costa et al. [99] Robak J, Shridi F, Wolbís M, Królikowska M. (1988) Screening of the influence of flavonoids on lipoxygenase and cyclooxygenase activity, as well as on nonenzymic lipid oxidation. Polish Journal of Pharmacology and Pharmacy, 40, 451-458. [100] Faizi S, Siddiqi H, Bano S, Naz A, Lubna, Mazhar K, Nasim S, Riaz T, Kamal S, Ahmad A, Khan AS. (2008) Antibacterial and antifungal activities of different parts of Tagetes patula: preparation of patuletin derivatives. Pharmaceutical Biology, 46, 309-320. [101] Maleki-Dizaji N, Fathiazad F, Garjani A. (2007) Antinociceptive properties of extracts and two flavonoids isolated from leaves of Danae racemosa. Archives of Pharmacal Research, 30, 1536-1542. [102] López-Sánchez C, Martín-Romero FJ, Sun F, Luis L, Samhan-Arias AK, García-Martínez V, Gutiérrez-Merino C. (2007) Blood micromolar concentrations of kaempferol afford protection against ischemia/reperfusion-induced damage in rat brain. Brain Research, 1182, 123-137. [103] Tu Y-C, Lian T-W, Yen J-H, Chen Z-T, Wu M-J. (2007) Antiatherogenic effects of kaempferol and rhamnocitrin. Journal of Agricultural and Food Chemistry, 55, 9969-9976. [104] Bestwick CS, Milne L, Duthie SJ. (2007) Kaempferol induced inhibition of HL-60 cell growth results from a heterogeneous response, dominated by cell cycle alterations. Chemico-Biological Interactions, 170, 76-85. [105] Leung HWC, Lin CJ, Hour MJ, Yang WH, Wang MY, Lee HZ. (2007) Kaempferol induces apoptosis in human lung non-small carcinoma cells accompanied by an induction of antioxidant enzymes. Food and Chemical Toxicology, 45, 2005-2013. [106] Hyun SK, Jung HA , Chung HY, Choi JS. (2006) In vitro peroxynitrite scavenging activity of 6-hydroxykynurenic acid and other flavonoids from Gingko biloba yellow leaves. Archives of Pharmacal Research, 29, 1074-1079. [107] Morikawa K, Nonaka M, Narahara M, Torii I, Kawaguchi K, Yoshikawa T, Kumazawa Y, Morikawa S. (2003) Inhibitory effect of quercetin on carrageenan-induced inflammation in rats. Life Sciences, 74, 709-721. [108] Abe F, Nagao T, Okabe H. (2002) Antiproliferative constituents in plants 9. Aerial parts of Lippia dulcis and Lippia canescens. Biological & Pharmaceutical Bulletin, 25, 920-922. [109] Miura K, Kikuzaki H, Nakatani N. (2002) Antioxidant activity of chemical components from sage (Salvia officinalis L.) and thyme (Thymus vulgaris L.) measured by the oil stability index method. Journal of Agricultural and Food Chemistry, 50, 1845-1851. [110] Steffen Y, Gruber C , Schewe T, Sies H. (2008) Mono-O-methylated flavanols and other flavonoids as inhibitors of endothelial NADPH oxidase. Archives of Biochemistry and Biophysics, 469, 209-219. [111] Xu YC, Leung SW, Yeung DK, Hu LH, Chen GH, Che CM, Man RY. (2007) Structure-activity relationships of flavonoids for vascular relaxation in porcine coronary artery. Phytochemistry, 68, 1179-1188. [112] Pelzer LE, Guardia T, Juarez AO, Guerreiro E. (1998) Acute and chronic antiinflammatory effects of plant flavonoids. Il Farmaco, 53, 421-424. [113] Kim M-J, Kim D-H, Lee KW, Yoon D-Y, Surh Y-J. (2007) Jaceosidin induces apoptosis in ras-transformed human breast epithelial cells through generation of reactive oxygen species. Annals of the New York Academy of Sciences, 1095, 483-495. [114] Lee SH, Bae E-A, Park E-K, Shin Y-W, Baek N-I, Han E-J, Chung H-G, Kim D-H. (2007) Inhibitory effect of eupatilin and jaceosidin isolated from Artemisia princeps in IgE-induced hypersensitivity. International Immunopharmacology, 7, 1678-1684. [115] Aron PM, Kennedy JA. (2008) Flavan-3-ols: nature, occurrence and biological activity. Molecular Nutrition & Food Research, 52, 79-104. [116] Rivière C, Richard T, Vitrac X, Merillon J-M, Valls J, Monti J-P. (2008) New polyphenols active on beta-amyloid aggregation. Bioorganic & Medicinal Chemistry Letters, 18, 828-831. [117] Lee SJ, Choi JH, Son KH, Chang HW, Kang SS, Kim HP. (1995) Suppression of mouse lymphocyte proliferation in vitro by naturally-occurring biflavonoids. Life Sciences, 57, 551-558. [118] Cherng J-M, Chiang W, Chiang L-C. (2008) Immunomodulatory activities of common vegetables and spices of Umbelliferae and its related coumarins and flavonoids. Food Chemistry, 106, 944-950. [119] Mookerjee BK, Lee TP, Lippes HA, Middleton E Jr. (1986) Some effects of flavonoids on lymphocyte proliferative responses. Journal of Immunopharmacology, 8, 371-392. Natural Product Communications Vol. 3 (12) 2008 2165 Natural Product Communications Manuscripts in Press Volume 3, Number 12 (2008) Characteristic Flavonoids from Acacia burkittii and A. acuminata Heartwoods and their Differential Cytotoxicity to Normal and Leukemia Cells Mary H. Grace, George R. Wilson, Fayez E. Kandil, Eugene Dimitriadis and Robert M. Coates Bioassay-Guided Isolation of Antiproliferative Compounds from Grape (Vitis vinifera) Stems Vincenzo Amico, Vincenza Barresi, Rosa Chillemi, Daniele Filippo Condorelli, Sebastiano Sciuto, Carmela Spatafora and Corrado Tringali Content of Total Carotenoids in Calendula officinalis L. from Different Countries Cultivated in Estonia Ain Raal, Kadri Kirsipuu, Reelika Must and Silvi Tenno Plant Growth Regulating Activity of Three Polyacetylenes from Helianthus annuus L. Si Won Hong, Koji Hasegawa, and Hideyuki Shigemori Bioactive Complex Triterpenoid Saponins Leguminosae Family José P. Parente and Bernadete P. da Silva from the Effects of the Essential Oil from Leaves of Alpinia zerumbet on Behavioral Alterations in Mice Shio Murakami, Mariko Matsuura, Tadaaki Satou, Shinichiro Hayashi and Kazuo Koike Discovering Protein Kinase C Active Plants Growing in Finland Utilizing Automated Bioassay Combined to LC/MS Anna Galkin, Jouni Jokela, Matti Wahlsten, Päivi Tammela, Kaarina Sivonen and Pia Vuorela Chemical Composition and Antimicrobial Activity of the Essential Oil from Chaerophyllum aureum L. (Apiaceae) Branislava Lakušić, Violeta Slavkovska, Milica Pavlović, Marina Milenković, Jelena Antić Stankovićc and Maria Couladis Isoprenylated Flavanones and Dihydrochalcones from Macaranga trichocarpa Yana M. Syah, Euis H. Hakim, Sjamsul A. Achmad, Muhamad Hanafi and Emilio L. Ghisalberti Abietane Diterpeniods from Hyptis verticillata Roy B. R. Porter, Duanne A. C. Biggs and William F. Reynolds Flavone and Flavonol Glycosides from Astragalus eremophilus and Astragalus vogelii Angela Perrone, Milena Masullo, Alberto Plaza, Arafa Hamed and Sonia Piacente Synthesis of Daldinol and Nodulisporin A by Oxidative Dimerization of 8-Methoxynaphthalen-1-ol Karsten Krohn and Abdulselam Aslan Diterpenoid Alkaloids from Aconitum longzhoushanense Ping He, Xi-Xian Jian, Dong-lin Chen and Feng-Peng Wang Alkaloids from Lindera aggregata Li-She Gan, Wei Yao, and Chang-Xin Zhou Antioxidants from the Leaf Extract of Byrsonima bucidaefolia G. Margarita Castillo-Avila, Karlina García-Sosa and Luis M. Peña-Rodríguez Salt Stress Induces Production of Melanin Related Metabolites in the Phytopathogenic Fungus Leptosphaeria maculans M. Soledade C. Pedras and Yang Yu Occurrence of Sesquiterpene Derivatives in Scleria striatonux De Wild (Cyperaceae) Kennedy Nyongbela, Karine Ioset Ndjoko, Reto Brun, Sergio Wittlin, James Mbah, Felix Makolo, Marc Akam, Claire Wirmum, Simon Efange and Kurt Hostettmann Cytotoxic Activity of Cycloartane Triterpenoids from Sphaerophysa salsula Dan Wang and Zhongjun Ma GC-MS Analysis and Antimicrobial Activity of Essential Oil of Stachys cretica subsp. smyrnaea Mehmet Öztürk, Mehmet Emin Duru, Fatma Aydoğmuş-Öztürk, Mansur Harmandar, Melda Mahlıçlı, Ufuk Kolak and Ayhan Ulubelen Antifungal and Insecticidal Activity of two Juniperus Essential Oils David E. Wedge, Nurhayat Tabanca, Blair J. Sampson, Christopher Werle, Betul Demirci, K. Husnu Can Baser, Peng Nan, Jian Duan and Zhijun Liu Minor Chemical Constituents of Vitex pinnata Athar Ata, Nathan Mbong, Chad D. Iverson and Radhika Samarasekera Essential Oil Compositions of Three Lantana Species from Monteverde, Costa Rica Ashley B. Walden, William A. Haber and William N. Setzer Design and Optimization of Ultrasound Assisted Extraction of Curcumin as an Effective Alternative for Conventional Solid Liquid Extraction of Natural Products Vivekananda Mandal, Saikat Dewanjee, Ranabir Sahu and Subhash C. Mandal Radical Scavenging Activity and Total Phenolic Content of Extracts of the Root Bark of Osyris lanceolata Elizabeth M. O. Yeboah and Runner R. T. Majinda Chemical Transformations of Parthenin, a Natural Bioactive Sesquiterpenoid Biswanath Das, G. Satyalakshmi, Nisith Bhunia, K. Ravider Reddy, V. Saidi Reddy and G. Mahender Cytotoxicity, Antimicrobial Activity and Composition of Essential Oil from Tanacetum balsamita L.subsp. balsamita Morteza Yousefzadi, Samad Nejad Ebrahimi, Ali Sonboli, Farah Miraghasi, Shahla Ghiasi, Mitra Arman and Nariman Mosaffa Betaines in Four Additional Phyla of Green Plants Gerald Blunden, Asmita Patel, Maricela Adrian Romero and Michael D. Guiry Natural Product Communications 2008 Volume 3 Natural Product Communications 3 (1-12) 1-2166 (2008) ISSN 1934-578X (print) ISSN 1555-9475 (online) NPC Natural Product Communications EDITOR-IN-CHIEF DR. PAWAN K AGRAWAL Natural Product Inc. 7963, Anderson Park Lane, Westerville, Ohio 43081, USA agrawal@naturalproduct.us EDITORS PROFESSOR GERALD BLUNDEN The School of Pharmacy & Biomedical Sciences, University of Portsmouth, Portsmouth, PO1 2DT U.K. axuf64@dsl.pipex.com PROFESSOR ALESSANDRA BRACA Dipartimento di Chimica Bioorganicae Biofarmacia, Universita di Pisa, via Bonanno 33, 56126 Pisa, Italy braca@farm.unipi.it PROFESSOR DEAN GUO State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100083, China gda5958@163.com PROFESSOR J. ALBERTO MARCO Departamento de Quimica Organica, Universidade de Valencia, E-46100 Burjassot, Valencia, Spain alberto.marco@uv.es PROFESSOR YOSHIHIRO MIMAKI School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Horinouchi 1432-1, Hachioji, Tokyo 192-0392, Japan mimakiy@ps.toyaku.ac.jp PROFESSOR STEPHEN G. PYNE Department of Chemistry University of Wollongong Wollongong, New South Wales, 2522, Australia spyne@uow.edu.au PROFESSOR MANFRED G. REINECKE Department of Chemistry, Texas Christian University, Forts Worth, TX 76129, USA m.reinecke@tcu.edu PROFESSOR WILLIAM N. SETZER Department of Chemistry The University of Alabama in Huntsville Huntsville, AL 35809, USA wsetzer@chemistry.uah.edu PROFESSOR YASUHIRO TEZUKA Institute of Natural Medicine Institute of Natural Medicine, University of Toyama, 2630-Sugitani, Toyama 930-0194, Japan tezuka@inm.u-toyama.ac.jp ADVISORY BOARD Prof. Viqar Uddin Ahmad Karachi, Pakistan Prof. Øyvind M. Andersen Bergen, Norway Prof. Giovanni Appendino Novara, Italy Prof. Yoshinori Asakawa Tokushima, Japan Prof. Maurizio Bruno Palermo, Italy Prof. Carlos Cerda-Garcia-Rojas Mexico city, Mexico Prof. Josep Coll Barcelona, Spain Prof. Geoffrey Cordell Chicago, IL, USA Prof. Samuel Danishefsky New York, NY, USA Dr. Biswanath Das Hyderabad, India Prof. A.A. Leslie Gunatilaka Tucson, AZ, USA Prof. Stephen Hanessian Montreal, Canada Prof. Michael Heinrich London, UK Prof. Kurt Hostettmann Lausanne, Switzerland Prof. Martin A. Iglesias Arteaga Mexico, D. F, Mexico Prof. Jerzy Jaroszewski Copenhagen, Denmark Prof. Teodoro Kaufman Rosario, Argentina Prof. Norbert De Kimpe Gent, Belgium Prof. Hartmut Laatsch Gottingen, Germany Prof. Marie Lacaille-Dubois Dijon, France Prof. Shoei-Sheng Lee Taipei, Taiwan Prof. Francisco Macias Cadiz, Spain Prof. Anita Marsaioli Campinas, Brazil Prof. Imre Mathe Szeged, Hungary Prof. Joseph Michael Johannesburg, South Africa Prof. Ermino Murano Trieste, Italy Prof. Virinder Parmar Delhi, India Prof. Luc Pieters Antwerp, Belgium Prof. Om Prakash Manhattan, KS, USA Prof. Peter Proksch Düsseldorf, Germany Prof. William Reynolds Toronto, Canada Prof. Raffaele Riccio Salerno, Italy Prof. Ricardo Riguera Santiago de Compostela, Spain Prof. Satyajit Sarker Coleraine, UK Prof. Monique Simmonds Richmond, UK Prof. Valentin Stonik Vladivostok, Russia Prof. Hermann Stuppner Innsbruck, Austria Prof. Apichart Suksamrarn Bangkock, Thailand Prof. Hiromitsu Takayama Chiba, Japan Prof. Karin Valant-Vetschera Vienna, Austria Prof. Peter G. Waterman Lismore, Australia Prof. Paul Wender Stanford, USA INFORMATION FOR AUTHORS Full details of how to submit a manuscript for publication in Natural Product Communications are given in Information for Authors on our Web site http://www.naturalproduct.us. Authors may reproduce/republish portions of their published contribution without seeking permission from NPC, provided that any such republication is accompanied by an acknowledgment (original citation)-Reproduced by permission of Natural Product Communications. Any unauthorized reproduction, transmission or storage may result in either civil or criminal liability. The publication of each of the articles contained herein is protected by copyright. Except as allowed under national “fair use” laws, copying is not permitted by any means or for any purpose, such as for distribution to any third party (whether by sale, loan, gift, or otherwise); as agent (express or implied) of any third party; for purposes of advertising or promotion; or to create collective or derivative works. Such permission requests, or other inquiries, should be addressed to the Natural Product Inc. (NPI). A photocopy license is available from the NPI for institutional subscribers that need to make multiple copies of single articles for internal study or research purposes. To Subscribe: Natural Product Communications is a journal published monthly. 2008 subscription price: US$1,395 (Print, ISSN# 1934-578X); US$1,095 (Web edition, ISSN# 1555-9475); US$1,795 (Print + single site online). Orders should be addressed to Subscription Department, Natural Product Communications, Natural Product Inc., 7963 Anderson Park Lane, Westerville, Ohio 43081, USA. Subscriptions are renewed on an annual basis. Claims for nonreceipt of issues will be honored if made within three months of publication of the issue. All issues are dispatched by airmail throughout the world, excluding the USA and Canada. Natural Product Communications Contents of Volume 3 2008 Number 1 A Novel Sesquiterpene from Pulicaria crispa (Forssk.) Oliv. Michael Stavri, Koyippally T. Mathew and Simon Gibbons 1 Cassane diterpenoids from Lonchocarpus laxiflorus John O. Igoli, Samuel O. Onyiriuka, Matthias C. Letzel, Martin N. Nwaji and Alexander I. Gray 5 COX-2 Inhibitory Activity of Cafestol and Analogs from Coffee Beans Ilias Muhammad, Satoshi Takamatsu, Jamal Mustafa, Shabana I. Khan, Ikhlas A. Khan, Volodymyr Samoylenko, Jaber S. Mossa, Farouk S. El-Feraly and D. Chuck Dunbar 11 Antibacterial Diterpenes from the Roots of Ceriops tagal Musa Chacha, Renameditswe Mapitse, Anthony J. Afolayan and Runner R. T. Majinda 17 Boswellic Acids with Acetylcholinesterase Inhibitory Properties from Frankincense Masahiro Ota and Peter J. Houghton 21 Synthesis of Pregnenolone and Methyl Lithocholate Oxalate Derivatives Lutfun Nahar, Satyajit D. Sarker and Alan B. Turner 27 Annona muricata (Graviola): Toxic or Therapeutic Sambeet Mohanty, Jackie Hollinshead, Laurence Jones, Paul Wyn Jones, David Thomas, Alison A. Watson, David G. Watson, Alexander I. Gray, Russell J. Molyneux and Robert J. Nash 31 Two New Alkylated Piperidine Alkaloids from Western Honey Mesquite: Prosopis glandulosa Torr. var. torreyana Volodymyr Samoylenko, D. Chuck Dunbar, Melissa R. Jacob, Vaishali C. Joshi, Mohammad K. Ashfaq and Ilias Muhammad 35 Selective Metabolism of Glycosidase Inhibitors by a Specialized Moth Feeding on Hyacinthoides non-scripta Flowers Alison A. Watson, Ana L. Winters, Sarah A. Corbet, Catherine Tiley and Robert J. Nash 41 Antimicrobial Activities of Alkaloids and Lignans from Zanthoxylum budrunga M. Mukhlesur Rahman, Alexander I. Gray, Proma Khondkar and M. Anwarul Islam 45 A Pyranochalcone and Prenylflavanones from Tephrosia pulcherrima (Baker) Drumm Seru Ganapaty, GuttulaV.K. Srilakshmi, Steve T. Pannakal and Hartmut Laatsch 49 Phenolic Glycosides from Phlomis lanceolata (Lamiaceae) Hossein Nazemiyeh, Abbas Delazar, Mohammed-Ali Ghahramani, Amir-Hossein Talebpour, Lutfun Nahar and Satyajit D. Sarker 53 Bisresorcinols and Arbutin Derivatives from Grevillea banksii R. Br. Hao Wang, David Leach, Michael C. Thomas, Stephen J. Blanksby, Paul I. Forster and Peter G. Waterman 57 Antioxidant and Membrane Stabilizing Properties of the Flowering Tops of Anthocephalus cadamba M. Ashraful Alam, Abdul Ghani, Nusrat Subhan, M. Mostafizur Rahman, M. Shamsul Haque, Muntasir M. Majumder, M. Ehsanul H. Majumder, Raushan A. Akter, Lutfun Nahar and Satyajit D. Sarker 65 A Method of Selecting Plants with Anti-inflammatory Potential for Pharmacological Study G. David Lin, Rachel W. Li, Stephen P. Myers and David N. Leach 71 Recent Advances of Biologically Active Substances from the Marchantiophyta Yoshinori Asakawa 77 Non-Protein Amino Acids: A Review of the Biosynthesis and Taxonomic Significance E. Arthur Bell (the late), Alison A. Watson and Robert J. Nash 93 Number 2 New cis-Chrysanthenyl Esters from Eryngium planum L. Emilia Korbel, Ange Bighelli, Anna Kurowska, Danuta Kalemba and Joseph Casanova 113 ii Contents of Volume 3 (1-12) 2008 Secondary Metabolites from Eremostachys laciniata İhsan Çalış, Ayşegül Güvenç, Metin Armağan, Mehmet Koyuncu, Charlotte H. Gotfredsen and Søren R. Jensen 117 A Novel Iridoid from Plumeria obtusa Firdous Imran Ali, Imran Ali Hashmi and Bina Shaheen Siddiqui 125 Terpenoids from Neolitsea dealbata Xiujun Wu, Bernhard Vogler, Betsy R. Jackes and William N. Setzer 129 Volatile Components from Selected Mexican, Ecuadorian, Greek, German and Japanese Liverworts Agnieszka Ludwiczuk, Fumihiro Nagashima, Rob S. Gradstein and Yoshinori Asakawa 133 New ent–Kaurane type Diterpene Glycoside, Pulicaroside-B, from Pulicaria undulata L. Nasir Rasool, Viqar Uddin Ahmad, Naseem Shahzad, Muhammad A. Rashid, Aman Ullah, Zahid Hassan, Muhammad Zubair and Rasool Bakhsh Tareen 141 Anti-babesial Quassinoids from the Fruits of Brucea javanica Ahmed Elkhateeb, Masahiro Yamasaki, Yoshimitsu Maede, Ken Katakura, Kensuke Nabeta and Hideyuki Matsuura 145 Triterpenoids and Alkaloids from the Roots of Peganum nigellastrum Zhongze Ma, Yoshio Hano, Feng Qiu, Gang Shao, Yingjie Chen and Taro Nomura 149 Saikosaponins from Bupleurum chinense and Inhibition of HBV DNA Replication Activity Feng Yin, Ruixiang Pan, Rongmin Chen and Lihong Hu 155 Brauhenoside A and B: Saponins from Stocksia brauhica Benth. Viqar Uddin Ahmad, Sadia Bader, Saima Arshad, Faryal Vali Mohammad, Amir Ahmed, Shazia Iqbal, Saleha Suleman Khan and Rasool Bakhsh Tareen 159 Saponins from Fresh Fruits of Randia siamensis (Lour) Roem. & Schult. (Rubiaceae) Rapheeporn Khwanchuea, Emerson Ferreira Queiroz, Andrew Marston, Chaweewan Jansakul and Kurt Hostettmann 163 New Alkaloid from Aspidosperma polyneuron Roots Tatiane Alves dos Santos, Dalva Trevisan Ferreira, Jurandir Pereira Pinto, Milton Faccione and Raimundo Braz-Filho 171 Acanthomine A, a new Pyrimidine-β-Carboline Alkaloid from the Sponge Acanthostrongylophora ingens Sabrin R. M. Ibrahim, RuAngelie Ebel, Rainer Ebel and Peter Proksch 175 Phytochemical and Microscopic Characterization of the Caribbean Aphrodisiac Bois Bandé: Two New Norneolignans Ingrid Werner, Pavel Mucaji, Armin Presser, Christa Kletter and Sabine Glasl 179 3-Acetoxy-7-methoxyflavone, a Novel Flavonoid from the Anxiolytic Extract of Salvia elegans (Lamiaceae) Silvia Marquina, Yolanda García, Laura Alvarez and Jaime Tortoriello 185 Struthiolanone: A Flavanone-Resveratrol Adduct from Struthiola argentea Sloan Ayers, Deborah L. Zink, Robert Brand, Seef Pretorius, Dennis Stevenson and Sheo B. Singh 189 New Acylated Flavonol Diglycosides of Cynanchum acutum Mona A. Mohamed, Wafaa S. Ahamed, Mortada M. El-Said and Heiko Hayen 193 Phenolic Constituents of Platanus orientalis L. Leaves Taha S. El-Alfy, Hamida M.A. El-Gohary, Nadia M. Sokkar, Amani A. Sleem and Dalia A. Al-Mahdy 199 Strepsiamide A-C, New Ceramides from the Marine Sponge Strepsichordaia lendenfeldi Sabrin R. M. Ibrahim, Gamal A. Mohamed, Ehab S. Elkhayat, Yaser G. Gouda and Peter Proksch 205 Free Radical Scavenging and Cytoprotective Activity of Salacia euphlebia Merr. Sanan Subhadhirasakul, Niwat Keawpradub, Charuporn Promwong and Supreeya Yuenyongsawad 211 Antialactone: A New γ-Lactone from Antiaris africana, and its Absolute Configuration Determined Vouffo Bertin, Hidayat Hussain, Simeon F. Kouam, Etienne Dongo, Gennaro Pescitelli, Piero Salvadori, Tibor Kurtán and Karsten Krohn 215 Subereaphenol A, a new Cytotoxic and Antimicrobial Dibrominated Phenol from the Red Sea Sponge Suberea mollis Lamiaa A. Shaala, Sherief I. Khalifa, Mostafa K. Mesbah, Rob W. M. van Soest and Diaa T. A. Youssef 219 A New Ferulic Ester and Related Compounds from Bombax malabaricum DC. Pahup Singh, Durga K. Mewara and Mahesh C. Sharma 223 Role of Turmeric in Ultraviolet Induced Genotoxicity in a Bacterial System Arijit Pal, Mita Ghosh and Arun Kumar Pal 227 Excited-State pKa Values of Curcumin Qian Zhao, De-Xin Kong and Hong-Yu Zhang 229 Antibacterial and Antifungal Activities of Some Phenolic Metabolites Isolated from the Lichenized Ascomycete Ramalina lacera Lumír O Hanuš, Marina Temina and Valery M Dembitsky 233 Contents of Volume 3 (1-12) 2008 iii Phenolic Constituents of Hypericum Flowers Carolina Nör, Ana Paula Machado Bernardi, Juliana Schulte Haas, Jan Schripsema, Sandra Beatriz Rech and Gilsane Lino von Poser 237 Seasonal Variation of Hypericin and Pseudohypericin Contents in Hypericum scabrum L. Growing Wild in Turkey Ali Kemal Ayan, Cüneyt Çırak and Kerim Güney 241 Molluscicidal Polyphenols from Species of Fucaceae Asmita V. Patel, David C. Wright, Maricela Adrian Romero, Gerald Blunden and Michael D. Guiry 245 Anti-diabetic Activity of Triphala Fruit Extracts, Individually and in Combination, in a Rat Model of Insulin Resistance Venkateshan S. Prativadibhayankaram, Samir Malhotra, Promila Pandhi and Amritpal Singh 251 Biotransformation of Mefenamic Acid by Cell Suspension Cultures of Solanum mammosum Suzana Surodjo, Angela A. Salim, Suciati, Achmad Syahrani, Gunawan Indrayanto and Mary J. Garson 257 Natural Variability in Enantiomeric Composition of Bioactive Chiral Terpenoids in the Essential Oil of Solidago canadensis L. from Uttarakhand, India Chandan S. Chanotiya and Anju Yadav 263 Germacrone Dominates the Leaf Oil of Siparuna grandiflora from Monteverde, Costa Rica William N. Setzer, Brittany R. Agius, Tameka M. Walker, Debra M. Moriarity and William A. Haber 267 Leaf Oil Composition of Piper aduncum subsp. ossanum (C. CD.) Saralegui from Cuba Orlando Abreu and Jorge A. Pino 271 Volatile Constituents from the Leaves of Phyllanthus salviaefolius H. B. K. from the Venezuelan Andes Silvana Villarreal, Luis B. Rojas, Alfredo Usubillaga, Irama Ramírez and Mariana Solórzano 275 Synergistic Antifungal Activities of Thymol Analogues with Propolis Chi-Pien Chen and Ai-Yu Shen 279 Argan oil, Functional Food, and the Sustainable Development of the Argan Forest Zoubida Charrouf and Dominique Guillaume 283 Chemical Constituents of Selected Japanese and New Zealand Liverworts Yoshinori Asakawa, Masao Toyota, Fumihiro Nagashima and Toshihiro Hashimoto 289 Number 3 Synthesis and Antibacterial Activity of Highly Oxygenated 1,8-Cineole Derivatives Margarita B. Villecco, Julieta V. Catalán, Marta I. Vega, Francisco M. Garibotto, Ricardo D. Enriz and César A. N. Catalán 303 Structure – Activity Relationships of Modified Eremophilanes and Anti-inflammatory Activity using the TPA Mouse Edema Ear Test Noemi Acevedo-Quiroz, Valeri Domínguez-Villegas and María Luisa Garduño-Ramírez, 313 New 3,4-Seco ent-Kaurenes from Croton caracasana Flowers Alírica I. Suárez, Katiuska Chavez, Franco Delle Monache, Luis Vasquez, Daniela M. Delannoy, Giovannina Orsini and Reinaldo S. Compagnone 319 Myrsicorianol, A New Prenylated Benzoic Acid Derivative from Myrsine coriacea Juan Manuel Amaro-Luis, Sonia Koteich-Khatib, Freddy Carrillo-Rodríguez and Alí Bahsas 323 Diastereoselective Synthesis of β-Hydroxyketones Teresa Mancilla Percino, Marisol López Martínez and José Luis León 329 Revised Structure by Computational Methods for a Coumarin Isolated from Zanthoxylum rhoifolium (Rutaceae) Augusto Rivera and Jaime Rios-Motta 333 1 13 Enantiodifferentiation by H and C NMR Spectroscopy (Dirhodium Method) – Selectivity of Oxygen Functionalities Edison Díaz Gómez, Sándor Antus, Renáta Ferenczi, Barbara Rys, Anna Stankiewicz and Helmut Duddeck 339 Antioxidant Activity of Fruits toward Iron under Gastrointestinal Conditions Helena Morais 345 Substrate Specificity of a Cationic Peptidase from Bromelia hemisphaerica L. Cortés-Vázquez Ma. Isabel, Muñoz-Sánchez José Luis and Briones-Martínez Roberto 351 New Sources of Oilseeds from Latin American Native Fruits Lilia Masson, Conrado Camilo, Katherine Gonzalez, Andrea Caceres, Neuza Jorge and Esperanza M. Torija 357 Experimental Design to Determine the Factors Affecting the Preparation of Extracts for Antibacterial Use Alejandro Pérez-López, Marcela Orozco-Hayek, Verónica Rivas-Galindo and Noemí Waksman de Torres 363 iv Contents of Volume 3 (1-12) 2008 Chemical Composition and Antibacterial Activity of the Essential Oil of Baccharis trinervis (Lam.) Pers. (Asteraceae) Collected in Venezuela Janne Rojas, Judith Velasco, Antonio Morales, Luis Rojas, Tulia Díaz, Maria Rondón and Juan Carmona 369 Supercritical Extraction of Essential Oil from Ilex paraguariensis Leaves Eduardo Cassel, Rubem M. F. Vargas and Gerti W. Brun 373 Cultivars of Lavandula lanata Boiss., a Good Source of Lavandulol Alejandro F. Barrero, M. Mar Herrador, Pilar Arteaga, Jesús F. Arteaga and Jesús Burillo 379 Essential Oils from Bolivia. X. Asteraceae: Gnaphalium viravira Molina Javier B Lopez Arze, Guy Collin, François-Xavier Garneau, France-Ida Jean and Hélène Gagnon 383 Synthesis of Sesquiterpenes via Silicon-Guided Rearrangement of Epoxydecalins Gonzalo Blay, Ana M. Collado, Begoña García and José R. Pedro 385 13 Structural Elucidation of Pimarane and Isopimarane Diterpenoids: The C NMR Contribution Ana M. L. Seca, Diana C. G. A. Pinto and Artur M. S. Silva 399 Anti-inflammatory and Cytotoxic Cycloartanes from Guayule (Parthenium argentatum) Graciela Flores-Rosete and Mariano Martínez-Vázquez 413 Rubus - A Review of its Phytochemical and Pharmacological Profile Guillermo Omar Rocabado, Luis Miguel Bedoya, María José Abad and Paulina Bermejo 423 Therapeutic Potential and Chemical Composition of Plants from the Genus Rubus: A Mini Review of the Last 10 Years Rivaldo Niero and Valdir Cechinel Filho 437 n Very Long-Range Correlations ( JC,H n > 3) in HMBC Spectra Ramiro Araya-Maturana, Hernán Pessoa-Mahana and Boris Weiss-López 445 Vibrational Circular Dichroism: A New Tool for the Solution-State Determination of the Structure and Absolute Configuration of Chiral Natural Product Molecules Laurence A. Nafie 451 Number 4 Screening Study of Potential Lead Compounds for Natural Product Based Fungicides from Juniperus lucayana Yarelis Ortíz Nuñez, Iraida Spengler Salabarria, Isidro G. Collado and Rosario Hernández-Galán 469 Inhibitory Activity of α,β-Unsaturated Lactones on Histamine Release from Rat Peritoneal Mast Cells Alicia B. Penissi, María I. Rudolph, Mariano E. Vera, María L. Mariani, Juan P. Ceñal, Carlos E. Tonn, Oscar S. Giordano and Ramón S. Piéis 475 Reactivity of Several Reactive Oxygen Species (ROS) with the Sesquiterpene Cacalol Manuel Jiménez-Estrada, Ricardo Reyes-Chilpa, Arturo Navarro-Ocaña and Daniel Arrieta-Báez 479 Ring Contraction of Gummiferolic Acid, a Diterpene Isolated from Margotia gummifera, Leading to Atisagibberellins Josefa Anaya, Juan José Fernández, Manuel Grande, Justo Martiáñez and Pascual Torres 483 A Convenient Synthesis of the Central Core of Helioporins, seco-Pseudopterosins and Pseudopterosins via BCA-Annulation Sequence Gema Esteban, Rocío Rincón, Aurelio G. Csákÿ and Joaquín Plumet 495 Role of Prostaglandins, Nitric oxide, Sulfhydryls and Capsaicin-sensitive Neurons in Gastroprotection of Stigmasterol and β-Sitosterol María Elena Sánchez-Mendoza, Jesús Arrieta and Andrés Navarrete 505 New Guanidine Alkaloids from the Leaves of Verbesina peraffinis Reinaldo S. Compagnone, Jhorman Bermudez, Glorymar Ibáñez, Beth Díaz, María R. Garrido, Anita Israel and Alírica I. Suárez 511 Aporphine Alkaloids from Guatteria stenopetala (Annonaceae) María Rodríguez, Elsy Bastidas, Mildred Rodríguez, Edgar Lucena, Anibal Castillo and Masahisa Hasegawa 515 Effects of Simple and Angular Chromones on Tumor Cell Respiration Ramiro Araya-Maturana, Jorge Heredia-Moya, Oscar Donoso-Tauda, Mario Vera, Jorge Toledo Hernández, Mario Pavani, Hernán Pessoa-Mahana, Boris Weiss-López and Jorge Ferreira 519 Effect on Hantavirus Replication of Resins from Heliotropium species and Other Selected Compounds René Torres Gaona, Héctor Galeno Araya and Brenda Modak Canobra 525 Total Synthesis of 5-(5–Ethenyl-2-Methoxyphenyl)–3-Furancarboxaldehyde and Related Compounds Leticia León–Galeana and Luis Ángel Maldonado G. 529 Contents of Volume 3 (1-12) 2008 v Seasonal Phytochemical Variation and Antifungal Evaluation of Different Parts of Epidendrum mosenii Patrícia Walter Rosa, Marina da Silva Machado, Rivaldo Niero, Susana Zacchino, Maria de los Ángeles Gette, Franco Delle Monache and Valdir Cechinel Filho 535 Acyl Sucroses from Salpichroa origanifolia Carmelo Dutra, María Verónica Cesio, Patrick Moyna and Horacio Heinzen 539 Influence of N-Deacetylation Conditions on Chitosan Production from α-Chitin Gemma Galed, Erika Diaz, Francisco M. Goycoolea and Angeles Heras 543 Searching for Natural Bioactive Compounds in Four Baccharis species from Bolivia Marcelo Dávila, Ingrid Loayza, Daniel Lorenzo and Eduardo Dellacassa 551 In vitro Antiprotozoal Activity and Chemical Composition of Ambrosia tenuifolia and A. scabra Essential Oils Valeria P. Sülsen, Silvia I. Cazorla, Fernanda M. Frank, Paola M. R. Di Leo Lira, Claudia A. Anesini, David Gutierrez Yapu, Alberto GiménezTurba, Arnaldo L. Bandoni, Emilio L. Malchiodi, Liliana V. Muschietti and Virginia S. Martino 557 Composition and Antioxidant activity of Essential Oils of Lippia origanoides H.B.K. grown in Colombia Elena Stashenko, Carlos Ruiz, Amner Muñoz, Martha Castañeda and Jairo Martínez 563 Germacrone: Occurrence, Synthesis, Chemical Transformations and Biological Properties Alejandro F. Barrero, M. Mar Herrador, Pilar Arteaga and Julieta V. Catalán 567 Terpenoids in Grapes and Wines: Origin and Micrometabolism during the Vinification Process Francisco M. Carrau, Eduardo Boido and Eduardo Dellacassa 577 Toxic Chemical Compounds of the Solanaceae Alicia B. Pomilio, Elvira M. Falzoni and Arturo A. Vitale 593 Synthesis of Marine Indole Alkaloids from Flustra foliacea Martha S. Morales-Ríos and Oscar R. Suárez-Castillo 629 Monoaminergic, Ion Channel and Enzyme Inhibitory Activities of Natural Aporphines, their Analogues and Derivatives Bruce K. Cassels and Marcelo Asencio 643 Number 5 Simultaneous Determination of Nine Bioactive Constituents of Caulis Lonicerae Japonicae by High-Performance Liquid Chromatography Coupled with Mass Spectrometry Hui-Jun Li, Zheng-Ming Qian, Ping Li , Mei-Ting Ren, Jun Chen and Gui-Zhong Xin 655 Comparative Analysis of Microbial and Rat Metabolism of the Total Saponins from Panax notoginseng by HPLC-ESI-MS/MS Guang-tong Chen, Min Yang, Si-jia Tao, Zhi-qiang Lu, Jin-qiang Zhang, Hui-lian Huang, Li-jun Wu and De-an Guo 661 Analysis on the Stability of Total Phenolic Acids and Salvianolic Acid B from Salvia miltiorrhiza by HPLC and HPLC-MSn Man Xu, Jian Han, Hui-feng Li, Li Fan, Ai-hua Liu and De-an Guo 669 Simultaneous Determination of Vitexin Rhamnoside and Vitexin Glucoside in Rats by Liquid Chromatography Coupled with Mass Spectrometry MingJin Liang, WeiDong Zhang, Chuan Zhang, YunHeng Shen, XiaoLin Wang and Xiangwei Wang 677 Identification and Quantification of three Tubeimosides in Rhizoma Bolbostematis by High Performance Liquid Chromatography with Evaporative Light Scattering Detection and Electrospray Mass Spectrometric Detection Hao Huang, MingJin Liang, Wen Xu, Chuan Zhang and WeiDong Zhang 683 Identification of the Major Constituents in Shi-Quan-Da-Bu Decoction by HPLC-ESI-MS/MS Sijia Tao, Guangtong Chen, Min Yang, Shaosheng Deng, Jinqiang Zhang and De-an Guo 689 Analysis of Major Chemical Constituents in Luan-Pao-Prescription Using Liquid Chromatography Coupled with Electrospray Ionization Mass Spectrometry Jin-qiang Zhang, Min Yang, Bao-hong Jiang, Hui-lian Huang, Guang-tong Chen, Zhi-qiang Lu, Xing-nuo Li, Kai-shun Bi and De-an Guo 697 Quantitative Determination of Ecdysteroids in Sida rhombifolia L. and various other Sida Species Using LC-UV, and their Anatomical Characterization Bharathi Avula, Vaishali Joshi, Yan-Hong Wang, Atul N. Jadhav and Ikhlas A. Khan 705 Antibacterial Effects of Guava Tannins and Related Polyphenols on Vibrio and Aeromonas Species Fumi Yamanaka, Tsutomu Hatano, Hideyuki Ito, Shoko Taniguchi, Eizo Takahashi and Keinosuke Okamoto 711 vi Contents of Volume 3 (1-12) 2008 Phytochemical Analysis of Nunavik Rhodiola rosea L. Vicky J. Filion, Ammar Saleem, Guy Rochefort, Marc Allard, Alain Cuerrier and John T. Arnason 721 1 Assessment of H NMR Spectroscopy for Specific Metabolite Fingerprinting of Angelica sinensis Dongmin Su, Jinglan Han, Shishan Yu and Hailin Qin 727 Steroidal Glycosides from the Seeds of Hyoscyamus niger L. Irina Lunga, Carla Bassarello, Pavel Kintia, Stepan Shvets, Sonia Piacente and Cosimo Pizza 731 A New Type of Modified Brassinosteroids for Enzyme-linked Immunosorbent Assay Vladimir Khripach, Vladimir Zhabinskii, Alexey Antonchick, Raissa Litvinovskaya, Svetlana Drach, Oleg Sviridov, Andrey Pryadko, Tatyana Novik, Vitaliy Matveentsev and Bernd Schneider 735 Puguflavanones A and B; Prenylated Flavanones from Baphia puguensis Modest C. Kapingu, Joseph J. Magadula, Zakaria H. Mbwambo and Dulcie A. Mulholland 749 New Flavonoids from Baeckea frutescens and their Antioxidant Activity Tran Hong Quang, Nguyen Xuan Cuong, Chau Van Minh, and Phan Van Kiem 755 Sideroxylin from Miconia ioneura: Monohydrate Crystal Structure from High Resolution X-Ray Powder Diffraction Silvina Pagola, María I. Tracanna, Sara M. Amani, Ana M. González, Ana B. Raschi, Elida Romano, Alba M. Benavente and Peter W. Stephens 759 Colletinin A and 2,2´´-Diepicolletinin A: Two New Bisflavan-3-ols from Rhododendron collettianum Viqar Uddin Ahmad, Farman Ullah, Hidayat Hussain, Gilles Dujardin, Arnaud Martel and Ivan Robert Green 765 Isoquinoline Alkaloids and Homoisoflavonoids from Drimiopsis barteri Bak and D. burkei Bak Dieudonne Ngamga, Josephine Bipa, Pearl Lebatha, Christabel Hiza, Joan Mutanyatta, Merha-Tibeb Bezabih, Pierre Tane and Berhanu M. Abegaz 769 Flavonoid Constituents of Carduncellus mareoticus (Del.) Hanelt and their Biological Activities Marwan M. Shabana, Moshera M. El-Sherei, Mohamed Y. Moussa, Amany A. Sleem and Hosam M. Abdallah 779 Isolation of Pyranocoumarins from Angelica gigas V.L. Niranjan Reddy, Atul N. Jadhav, Bharathi Avula and Ikhlas A. Khan 785 Constituents of Zanthoxylum flavum and their Antioxidant and Antimalarial Activities Samir A. Ross, Kesanapallis Krishnaven, Mohamed M. Radwan, Satoshi Takamatsu and Charles L. Burandt 791 Two New Xanthone Glycosides from Ventilago leiocarpa Benth. Li-Li Wang, Jian-Ping Zuo, Lei Ma, Xia-Chang Wang and Li-Hong Hu 795 New Derivatives of Chromene and Acetoxyeudesmane Obtained by Microbial Transformation Mamdouh S. A. Haridy and Abou El-Hamd H. Mohamed 799 Dihydrostilbenes from Indigofera pulchra Aliyu Musa, A. K. Haruna, M. Ilyas, Augustine Ahmadu, Simon Gibbons and M. Mukhlesur Rahman 805 Membrane Activity-Guided Isolation of Antiproliferative and Antiplatelet Constituent from Evodiopanax innovans Hironori Tsuchiya, Toshiyuki Tanaka, Motohiko Nagayama, Masayoshi Oyama and Munekazu Iinuma 809 O-Methylheptaphylline from Clausena suffruticosa Rokeya Begum, Mohammad S. Rahman, A. M. Sarwaruddin Chowdhury , M. Mukhlesur Rahman and Mohammad A. Rashid 815 The Anti-tumor Effect of the Light Petroleum Extract from Pulsatilla chinensis (Bunge) Regel Min Zhang, Zhihang Song, Dan Wang, Lin Cheng, Wenrong Jin, Peng Zhang, Yang Huo and Zhongjun Ma 819 Thymol and its Derivatives as Antimicrobial Agents Ajai Kumar, Suriya P. Singh and Sudarshan S. Chhokar 823 Seasonal Variation in the Leaf Essential Oil Composition of Sassafras albidum Kristi M. Kaler and William N. Setzer 829 Essential Oil Composition of Two Subspecies of Nepeta glomerulosa Boiss. from Iran Katayoun Javidnia, Ramin Miri, Shaghayegh Rasteh Rezazadeh, Mohammad Soltani and Ahmad Reza Khosravi 833 Antioxidant Activities of Eleven Australian Essential Oils Qian Zhao, Esther Joy Bowles and Hong-Yu Zhang 837 5α, 8α-Epidioxysterol from the marine sponge Biemna triraphis Topsent. Julia Bensemhoun, Isabelle Bombarda, Maurice Aknin, Jean Vacelet and Emile M. Gaydou 843 Number 6 Iridoid Glucosides from Viburnum macrocephalum Lamberto Tomassini, Sebastiano Foddai, Antonio Ventrone and Marcello Nicoletti 845 Contents of Volume 3 (1-12) 2008 vii Chemotaxonomy of Linaria Genus by Nor-Iridoids Distribution Mauro Serafini, Antonella Piccin, Alessandra Stanzione, Valentina Petitto and Marcello Nicoletti 847 A Guaiane Diol from Actinolema eryngioides Hasan Çetin Özen, Federica Pollastro and Giovanni Appendino 849 Anti-inflammatory Activity of New Guaiane Acid Derivatives from Achillea Coarctata Mohamed-Elamir F. Hegazy, Ahmed Abdel-Lateff, Amira M. Gamal-Eldeen, Fatma Turky, Toshifumi Hirata, Paul W. Paré, Joe Karchesy, Mohamed S. Kamel and Ahmed A. Ahmed 851 Chemical Constituents of Ximenia americana Mônica Regina Silva de Araújo, João Carlos da Costa Assunção, Ivana Nogueira Fernandes Dantas, Letícia Veras Costa-Lotufo and Francisco José Queiroz Monte 857 Isolation and Cytotoxic Activity of Compounds from the Root Tuber of Curcuma wenyujin Dan Wang, Wei Huang, Qiang Shi, Chengtao Hong, Yiyu Cheng, Zhongjun Ma and Haibin Qu 861 Cruzain Inhibition by Terpenoids: A Molecular Docking Analysis Ifedayo V. Ogungbe and William N. Setzer 865 A new 14,15-dinor-labdane Glucoside from Crassocephalum mannii Mohamed-Elamir F. Hegazy, Ashraf A. Aly, Ahmed A. Ahmed, Djemgou C. Pierre, Pierre Tane and Mehawed M. Ahmed 869 Furanolabdane Diterpene Glycosides from Eremostachys laciniata Abbas Delazar, Masoud Modarresi, Hossein Nazemiyeh, Fatemeh Fathi-Azad, Lutfun Nahar and Satyajit D. Sarker 873 Secondary Metabolites from the Aerial Parts of Salvia aethiopis L. Anna Malafronte, Fabrizio Dal Piaz, Giuseppina Cioffi , Alessandra Braca, Antonella Leone, and Nunziatina De Tommasi 877 Platelet Antiaggregating Activity and Chemical Constituents of Salvia x jamensis J. Compton Angela Bisio, Giovanni Romussi, Eleonora Russo, Nunziatina De Tommasi, Nicola Mascolo, Alessio Alfieri, Maria Carmela Bonito and Carla Cicala 881 Triterpenoids from Anthocleista grandiflora (Gentianaceae) Joseph J. Magadula, Dulcie A. Mulholland and Neil R. Crouch 885 An Immunomodulator from Terminalia arjuna and Biological Evaluation of its Derivatives Mohit Saxena, Sachidanand Yadav, Dhyaneshwar U. Bawankule, Santosh K. Srivastava, Anirban Pal, Rupal Mishra, Madan M. Gupta, Mahendra P. Darokar, Priyanka and Suman P. S. Khanuja 891 New Antiinflammatory Cycloart-23-ene-3β-ol from Senefelderopsis chibiriquetensis Dilsia J. Canelón, Alírica I. Suárez, Juan De Sanctis, Michael Mijares and Reinaldo S. Compagnone 895 New Cycloartane Glycosides from Camptosorus sibiricus Rupr. Peng Zhang, Yiyu Cheng and Zhongjun Ma 899 Triterpene Bisdesmosides from the Stems of Akebia quinata Yoshihiro Mimaki and Saya Doi 903 Chemotaxonomic relationships in Celastraceae inferred from Principal Component Analysis (PCA) and Partial Least Squares (PLS) Ana V. Mello Cruz, Marcelo J. P. Ferreira, Marcus T. Scotti, Maria A. C. Kaplan and Vicente P. Emerenciano 911 Chemical Composition of the Essential Oils of Centaurea sicana and C. giardinae Growing Wild in Sicily Carmen Formisano, Daniela Rigano, Felice Senatore, Maurizio Bruno, Sergio Rosselli, Francesco Maria Raimondo and Vivienne Spadaro 919 Chemical Composition of the Essential Oil of the Flowering Aerial Parts of Craniotome furcata Rajesh K. Joshi and Chitra Pande 923 Chemical Composition of the Essential Oils from Flower, Leaf, and Stem of Senecio trapezuntinus Boiss. Grown in Turkey Osman Üçüncü, Nuran Yaylı, Ahmet Yaşar, Salih Terzioğlu and Nurettin Yaylı 925 GC/MS Analysis of the Essential Oil from the Oleoresin of Pistacia atlantica Desf. subsp. Atlantica from Algeria Hachemi Benhassaini, Fatima Z. Bendeddouche, Zoheir Mehdadi and Abderrahmane Romane 929 Antibacterial Activity and Composition of the Essential Oil of Peperomia galioides HBK (Piperaceae) from Peru Vincenzo De Feo, Alberto Juarez Belaunde, Joe Guerrero Sandoval Felice Senatore and Carmen Formisano 933 Chemical Composition and in vitro Antibacterial Activity of the Essential Oil of Calycolpus moritzianus (O. Berg) Burret from Mérida, Venezuela Teresa Díaz, Flor D. Mora, Judith Velasco, Tulia Díaz, Luis B. Rojas, Alfredo Usubillaga and Juan Carmona A 937 Essential Oil Analysis and Antimicrobial Activity of Paeonia mascula from Turkey Nurettin Yaylı, Ahmet Yaşar, Nuran Yaylı, Mesut Albay and Kamil Coşkunçelebi 941 viii Contents of Volume 3 (1-12) 2008 Antimicrobial Activity of Nepeta Isolates Chandra S. Mathela and Neeta Joshi 945 Sesquiterpenes of Lactarius and Russula (Mushrooms): An Update Marco Clericuzio, Gianluca Gilardoni, Omar Malagòn, Giovanni Vidari and Paola Vita Finzi 951 Virescenols: Sources, Structures and Chemistry Maria Carla Marcotullio, Ornelio Rosati and Massimo Curini 975 Rearranged Clerodane and Abietane Derived Diterpenoids from American Salvia Species Baldomero Esquivel 989 Diterpenes from Euphorbia as Potential Leads for Drug Design Elisa Barile, Gabriella Corea and Virginia Lanzotti 1003 neo-Clerodane Diterpenoids from Verbenaceae: Structural Elucidation and Biological Activity Amaya Castro and Josep Coll 1021 Number 7 Chemical Composition, Olfactory Evaluation and Antioxidant Effects of the Essential Oil of Satureja montana L. Ivanka Stoilova, Stefanie Bail, Gerhard Buchbauer, Albert Krastanov, Albena Stoyanova, Erich Schmidt and Leopold Jirovetz 1035 Chemical Composition, Olfactory Evaluation and Antioxidant Effects of an Essential Oil of Origanum vulgare L. from Bosnia Ivanka Stoilova, Stefanie Bail, Gerhard Buchbauer, Albert Krastanov, Albena Stoyanova, Erich Schmidt and Leopold Jirovetz 1043 Chemical Composition, Olfactory Evaluation and Antioxidant Effects of an Essential Oil of Thymus vulgaris L. from Germany Ivanka Stoilova, Stefanie Bail, Gerhard Buchbauer, Albert Krastanov, Albena Stoyanova, Erich Schmidt and Leopold Jirovetz 1047 Chemical Composition, Olfactory Evaluation and Antioxidant Effects of the Essential oil of Origanum majorana L. from Albania Erich Schmidt, Stefanie Bail, Gerhard Buchbauer, Ivanka Stoilova, Albert Krastanov, Albena Stoyanova and Leopold Jirovetz 1051 GC-MS-Analysis, Antimicrobial Activities and Olfactory Evaluation of Essential Davana (Artemisia pallens Wall. ex DC) Oil from India Stefanie Bail, Gerhard Buchbauer, Erich Schmidt, Juergen Wanner, Alexander Slavchev, Albena Stoyanova, Zapriana Denkova, Margit Geissler and Leopold Jirovetz 1057 Comparative Evaluation of Antimicrobial Activity andComposition of Rose Oils From Various Geographic Origins, in Particular Bulgarian Rose Oil Velizar Gochev, Katrin Wlcek, Gerhard Buchbauer, Albena Stoyanova, Anna Dobreva, Erich Schmidt and Leopold Jirovetz 1063 Composition and Antimicrobial Analysis of the Essential Oil of Litsea laevigata Nees. (Lauraceae) Muhammed Arif M, Subbu Raj M, Leopold Jirovetz and Mohamed Shafi P 1069 Chemical Composition and Antifungal Activity of Angelica sinensis Essential Oil against three Colletotrichum species Nurhayat Tabanca, David E. Wedge, Xiaoning Wang, Betul Demirci, Kemal Husnu Can Baser, Ligang Zhou and Stephen J. Cutler 1073 Development of a Miniaturized 24-well Strawberry Leaf Disk Bioassay for Evaluating Natural Fungicides Xiaoning Wang, David E. Wedge, Nurhayat Tabanca, Robert D. Johnson, Stephen J. Cutler, Patrick F. Pace, Barbara J. Smith and Ligang Zhou 1079 Investigation of Anticancer and Antiviral Properties of Selected Aroma Samples Boris Ryabchenko, Elena Tulupova, Erich Schmidt, Katrin Wlcek, Gerhard Buchbauer and Leopold Jirovetz 1085 Chemical and Biological Investigations of Essential Oils from Stem Barks of Enantia chlorantha Oliv. and Polyalthia suaveolens Engler. & Diels. from Cameroon Maximilienne Nyegue, Paul-Henri Amvam-Zollo, François-Xavier Etoa, Huguette Agnaniet and Chantal Menut 1089 Evaluation of the Activities of Five Essential Oils against the Stored Maize Weevil Oluwakemi O. Odeyemi, Patrick Masika and Anthony J. Afolayan 1097 Physiological and Behavioral Effects of 1,8-Cineol and (±)-Linalool: A Comparison of Inhalation and Massage Aromatherapy Eva Heuberger, Josef Ilmberger, Engelbert Hartter and Gerhard Buchbauer 1103 Synergistic and Antagonistic Interactions of Essential Oils on the Biological Activities of the Solvent Extracts from Three Salvia species Guy P. P. Kamatou, Robyn L. van Zyl, Hajierah Davids, Sandy F. van Vuuren and Alvaro M. Viljoen 1111 Contents of Volume 3 (1-12) 2008 ix Essential Oil Analysis of the Follicles of Four North American Magnolia Species Wolfgang Schühly, Samir A. Ross, Zlatko Mehmedic and Nikolaus H. Fischer 1117 Essential Oil Composition of Eryngium campestre L. Growing in Different Soil Types. A Preliminary Study Jesús Palá-Paúl, Jaime Usano-Alemany, A. Cristina Soria, M. José Pérez-Alonso and Joseph J. Brophy 1121 Essential Oil Compounds of Origanum vulgare L. (Lamiaceae) from Corsica Brigitte Lukas, Corinna Schmiderer, Ulrike Mitteregger, Chlodwig Franz and Johannes Novak 1127 Comparative Study of the Chemical Profiles of the Essential Oils of Ripe and Rotten Fruits of Citrus aurantifolia Swingle Anthony J. Afolayan and Olayinka T. Asekun 1133 Chemical Variation in the Essential Oil Composition of Hyptis suaveolens (L.) Poit. (Lamiaceae) Paolo Grassi, Marvin José Nuñez, Tomás Sigfrido Urías Reyes and Chlodwig Franz 1137 Variability of the Volatile Oil Composition in a Population of Silaum silaus from Eastern Austria Remigius Chizzola 1141 The Bark Essential Oil Composition and Chemotaxonomical Appraisal of Cedrelopsis grevei H. Baillon from Madagascar Miarantsoa Rakotobe, Chantal Menut, Hanitriniaina Sahondra Andrianoelisoa, Voninavoko Rahajanirina, Philippe Collas de Chatelperron, Edmond Roger and Pascal Danthu 1145 Essential Oil Polymorphism of Hungarian Common Thyme (Thymus glabrescens Willd.) Populations Zsuzsanna Pluhár, Szilvia Sárosi, Ildikó Novák and Gabriella Kutta 1151 Diversity of Essential Oil Glands of Spanish Sage (Salvia lavandulifolia Vahl, Lamiaceae) Corinna Schmiderer, Paolo Grassi, Johannes Novak and Chlodwig Franz 1155 Micro-bore Column Fast Gas Chromatography-Mass Spectrometry in Essential Oil Analysis Peter Quinto Tranchida, Rosaria Costa, Paola Dugo, Giovanni Dugo and Luigi Mondello 1161 Carbon Isotope Ratio Analysis of Authentic and Commercial Essential Oils of Lemon Balm Susanne Wagner, Polona Vreca, Albrecht Leis and Herbert Boechzelt 1165 Essential Oil Compounds for Thrips Control – A Review Elisabeth H. Koschier 1171 Linalool – A Review of a Biologically Active Compound of Commercial Importance Guy P. P. Kamatou and Alvaro M. Viljoen 1183 Limonene - A Review: Biosynthetic, Ecological and Pharmacological Relevance Paul Erasto and Alvaro M.Viljoen 1193 Number 8 Plant Secondary Metabolism: Diversity, Function and its Evolution Michael Wink 1205 Major Constituents of the Predominant Endophytic Fungi from the Nigerian Plants Bryophyllum pinnatum, Morinda lucida and Jathropha gossypiifolia Abimbola A. Sowemimo, RuAngelie Edrada-Ebel, Rainer Ebel, Peter Proksch, Olanrewaju R. Omobuwajo and Saburi A. Adesanya 1217 Integration of Plasma Membrane and Nuclear Signaling in Elicitor Regulation of Plant Secondary Metabolism Gastón Stockman and Ricardo Boland 1223 Selective Elicitation of the Phytoalexin Rutalexin in Rutabaga and Turnip Roots by a Biotrophic Plant Pathogen M. Soledade C. Pedras, Ravi S. Gadagi, Qing-An Zheng and S. Roger Rimmer 1239 Transient Induction of Antimicrobial 3-Deoxyflavonoids does not affect Pharmacological Compounds in Hawthorn Karin Schlangen, Heidi Halbwirth, Silke Peterek, Christian Gosch, Alexandra Ringl, Thilo C. Fischer, Dieter Treutter, Gert Forkmann, Brigitte Kopp and Karl Stich 1245 Trafficking and Sequestration of Anthocyanins Erich Grotewold and Kevin Davies 1251 Unraveling the Biochemical Base of Dahlia Flower Coloration Heidi Halbwirth, Gerlinde Muster and Karl Stich 1259 Anthocyanins Reduce Fungal Growth in Fruits H. Martin Schaefer, Michael Rentzsch and Michael Breuer 1267 Phenolic Compounds Produced by Secretory Structures in Plants: a Brief Review Marilia de M. Castro and Diego Demarco 1273 x Contents of Volume 3 (1-12) 2008 Surface Flavonoids in Catalpa ovata, Greyia sutherlandii and Paulownia tomentosa Eckhard Wollenweber, Rüdiger Wehde, Matthias Christ and Marion Dörr 1285 Chemodiversity of Artemisia dracunculus L. from Kyrgyzstan: Isocoumarins, Coumarins, and Flavonoids from Aerial Parts Tshering D. Bhutia and Karin M. Valant-Vetschera 1289 Occurrence and Distribution of the Flavone Tricetin and its Methyl Derivatives as Free Aglycones Eckhard Wollenweber and Marion Dörr 1293 Variation in Flavonoids in Leaves, Stems and Flowers of White Clover Cultivars Sandra C.K. Carlsen, Anne G. Mortensen, Wieslaw Oleszek, Sonia Piacente, Anna Stochmal and Inge S. Fomsgaard 1299 Plants as a Still Unexploited Source of New Drugs Kurt Hostettmann and Andrew Marston 1307 Activity-guided Isolation and Identification of Free Radical-scavenging Components from Ethanolic Extract of Boneset (Leaves of Eupatorium perfoliatum) Solomon Habtemariam 1317 Activity-guided Isolation and Identification of Antioxidant Components from Ethanolic Extract of Peltiphyllum peltatum (Torr.) Engl. Solomon Habtemariam 1321 Two New Kaempferol Glycosides from Matthiola longipetala (subsp. livida) (Delile) Maire and Carcinogenic Evaluation of its Extract Mona M. Marzouk, Salwa A. Kawashty, Lamyaa F. Ibrahium, Nabiel A. M. Saleh and Abdel-Ssalam M. Al-Nowaihi 1325 Newer Constituents of Derris indica stem Thangaraj Shankar, Shanmugam Muthusubramanian and Rathinasamy Gandhidasan 1329 Structural Characterization of Genuine (–)-Pipermethystine, (–)-Epoxypipermethystine, (+)-Dihydromethysticin and Yangonin from the Kava Plant (Piper methysticum) Panče Naumov, Klaus Dragull, Masahiro Yoshioka, Chung-Shih Tang and Seik Weng Ng 1333 Naturally Occurring Flavanones: An Overview Goutam Brahmachari 1337 Bioisosteric Replacement of Molecular Scaffolds: From Natural Products to Synthetic Compounds Kristina Grabowski, Ewgenij Proschak, Karl-Heinz Baringhaus, Oliver Rau, Manfred Schubert-Zsilavecz and Gisbert Schneider 1355 Composition of Essential Oil from Lavandula angustifolia and L. intermedia VarietiesGrown in British Columbia, Canada W. Alexander Lane and Soheil S. Mahmoud 1361 Chemical Compositions and Biological Activities of Leaf Essential Oils of Twelve Species of Piper from Monteverde, Costa Rica William N. Setzer, Grace Park, Brittany R. Agius, Sean L. Stokes, Tameka M. Walker and William A. Haber 1367 Number 9 Anti-tuberculosis Compounds from two Bolivian Medicinal Plants, Senecio mathewsii and Usnea florida Qi Hong, David E. Minter, Scott G. Franzblau and Manfred G. Reinecke 1377 Natural Products from Polygonum perfoliatum and their Diverse Biological Activities Chi-I Chang, Fei-Jane Tsai and Chang-Hung Chou 1385 Phytochemical Characterization of the Australian (Aboriginal) Medicinal Plant Dolichandrone heterophylla and Influence of Selected Isolated Compounds on Human Keratinocytes Thomas Dzeha, Kristian Wende, Manuela Harms, Ju Ju (Burriwee) Wilson, Jim Kohen, Subra Vemulpad, Joanne Jamie and Ulrike Lindequist 1387 Analysis of Saponin Mixtures from Alfalfa (Medicago sativa L.) Roots using Mass Spectrometry with MALDI Techniques H. Ewa Witkowska, Zbigniew Bialy, Marian Jurzysta and George R. Waller 1395 Control of Allantoin Accumulation in Comfrey Paulo Mazzafera, Kátia Viviane Gonçalves and Milton Massao Shimizu 1411 Trigonelline (N-methylnicotinic acid) Biosynthesis and its Biological Role in Plants Hiroshi Ashihara 1423 Contents of Volume 3 (1-12) 2008 xi Biosynthesis and Catabolism of Purine Alkaloids in Camellia Plants Misako Kato and Hiroshi Ashihara 1429 A View on the Active Site of Firefly Luciferase Franklin R. Leach Study on the Chemical Constituents of Premna integrifolia L. Nguyen Thi Bich Hang, Pham Thanh Ky, Chau Van Minh, Nguyen Xuan Cuong, Nguyen Phuong Thao and Phan Van Kiem 1449 Τ-Cadinol Nerolidol Ether from Schisandra chinensis Asmita V. Patel, Gerald Blunden, Peter D. Cary, Lubomír Opletal, Markéta Beránkova, Kersti Karu, David E. Thurston and Milan Pour 1453 Bioactive Semisynthetic Derivatives of (S)-(+)-Curcuphenol Helena Gaspar, Cristina Moiteiro, João Sardinha and Azucena González-Coloma 1457 Eco-contribution to the Chemistry of Perezone, a Comparative Study, Using Different Modes of Activation and Solventless Conditions Joel Martínez, Benjamín Velasco-Bejarano, Francisco Delgado, Rocío Pozas, Héctor M. Torres Domínguez, José G. Trujillo Ferrara, Gabriel A. Arroyo and René Miranda 1465 Chemical Composition of Diterpenes from the Brown Alga Canistrocarpus cervicornis (Dictyotaceae, Phaeophyceae) Aline Santos de Oliveira, Diana Negrão Cavalcanti, Éverson Miguel Bianco, Joel Campos de Paula, Renato Crespo Pereira, Yocie Yoneshigue-Valentin and Valéria Laneuville Teixeira 1469 Cembranoid Diterpenes from the Soft Corals Sarcophyton sp. and Sarcophyton glaucum Daniela Grote, Kamel H. Shaker, Hesham S. M. Soliman, Muhammmad M. Hegazi and Karlheinz Seifert 1473 Dolabellane Diterpenes from Cleome droserifolia Hoda M. Fathy, Mohamed I. Aboushoer, Fathallah M. Harraz, Abdallah A. Omar, Gilles Goetz and Rafael Tabacchi 1479 Constituents of ‘Caincin’, a Bioactive Saponin Fraction from the Root-bark of Chiococca alba (L.) Hitch. Jnanabrata Bhattacharyya, S. K. Srivastava, M. F. Agra and George Majetich 1483 A New Biflavanone from Ochna lanceolata Shaik I. Khalivulla, Nimmanapalli P. Reddy, Bandi A.K. Reddy, Ramireddy V.N. Reddy, Duvvuru Gunasekar, Alain Blond and Bernard Bodo 1487 Isoflavonoids from an Egyptian Collection of Colutea istria Mohamed M. Radwan 1491 A Flavonoid Glycoside from the Leaves of Morinda tinctoria Atish K. Sahoo, Nisha Narayanan, S. Rajan and Pulok K. Mukherjee 1495 New Anthocyanins from Stem Bark of Castor, Ricinus communis Robert Byamukama, Monica Jordheim, Bernard Kiremire and Øyvind M. Andersen 1497 Prenylated Xanthones and a Benzophenone from Baphia kirkii Modest C. Kapingu and Joseph J. Magadula 1501 Phytochemical and Antimicrobial Investigation of Latex from Euphorbia abyssinica Gmel. Fathy EL- Fiky, Kaleab Asres, Simon Gibbons, Hala Hammoda, Jihan Badr and Shemsu Umer 1505 Fenugreek Extract Rich in 4-Hydroxyisoleucine and Trigonelline Activates PPARα and Inhibits LDL Oxidation: Key Mechanisms in Controlling the Metabolic Syndrome Alvin Ibarra, Kan He, Naisheng Bai, Antoine Bily, Marc Roller, Aurélie Coussaert, Nicolas Provost and Christophe Ripoll 1509 Comparison of Different Techniques for Extraction of Biologically Active Compounds from Achillea Millefolium Proa Antoaneta Trendafilova and Milka Todorova 1515 Extraction Methods Play a Critical Role in Chemical Profile and Biological Activities of Black Cohosh Bei Jiang, Kurt A. Reynertson, Amy C. Keller, Linda S. Einbond, Debra L. Bemis, I. Bernard Weinstein, Fredi Kronenberg and Edward J. Kennelly 1519 Antimicrobial Activity and Chemical Composition of Callistemon pinifolius and C. salignus Leaf Essential Oils from the Northern Plains of India Mohit Saxena, Kunal Shrivastava, Santosh K. Srivastava, Suaib Luqman, Ajai Kumar, Mahendra. P. Darokar, Kodakandla V. Syamsundar, Tota Ram and Suman P. S. Khanuja 1533 Analysis of Chemical Constituents of Tithonia rotundifolia Leaf Essential Oil Found in Nigeria Adebayo A. Gbolade, Vânia Tira-Picos and J.M.F. Nogueira 1537 Sedative Effect of Eucalyptus urophylla and E. brassiana in mice Gisele F.D. Teixeira, Roberto C.P. Lima Júnior, Edilberto R. Silveira, Marinalva O. Freitas and Adriana R. Campos 1539 1437 xii Contents of Volume 3 (1-12) 2008 In Vitro Antifungal Activity of Polysulfides-Rich Essential Oil of Ferula latisecta Fruits against Human Pathogenic Dermatophytes Mehrdad Iranshahi, Abdolmajid Fata, Bahareh Emami, Bibi Mohadeseh Jalalzadeh Shahri and Bibi Sedigheh Fazly Bazzaz 1543 Biological Activity and Composition of the Essential Oil of Dracocephalum moldavica L. Grown in Iran Ali Sonboli, Mehran Mojarrad, Abbas Gholipour, Samad Nejad Ebrahimi and Mitra Arman 1547 Antioxidant Activity and Chemical Composition of Essential Oils from Schinus fasciculata (Griseb.) I.M. Johnst and S. praecox (Griseb.) Speg. Ana P. Murray, Silvana A. Rodriguez and María G. Murray 1551 Essential oil Composition of Dendropanax gonatopodus from Monteverde, Costa Rica. An ab initio Examination of Aromadendrane Sesquiterpenoids William N. Setzer 1557 Number 10 Oxidative Transformations of Lappaconitine and 19-Oxolappaconine, Structural Revision of an obtained 8,9-Seco Product Elza U. Shafikova, Elena M. Tsyrlina, Leonid V. Spirikhin, Alsu A. Balandina, Shamil K. Latypov, Marat S. Yunusov and Oleg G. Sinyashin 1565 Oxidative Decarboxylation of Triterpene C-28 Acids of Lupane Series Nataliya G. Кomissarova, Nataliya G. Belenkova, Olga V. Shitikova, Leonid V. Spirikhin and Marat S. Yunusov 1569 Proapoptotic and Anticarcinogenic Activities of Leviusculoside G from the Starfish Henricia leviuscula and Probable Molecular Mechanism Sergey N. Fedorov, Larisa K. Shubina, Alla A. Kicha, Natalia V. Ivanchina, Jong Y. Kwak, Jun O. Jin, Ann M. Bode, Zigang Dong and Valentin A. Stonik 1575 Two New Steroid Oligoglycosides from the Caribbean Sponge Mycale laxissima Shamil Sh. Afiyatullov, Alexandr S. Antonov, Anatoly I. Kalinovsky and Pavel S. Dmitrenok 1581 New Polar Steroids from Starfish Valentin A. Stonik, Natalia V. Ivanchina and Alla A. Kicha 1587 New Angucyclinones from the Marine Mollusk Associated Actinomycete Saccharothrix espanaensis An 113 Nataliya I. Kalinovskaya, Anatoly I. Kalinovsky, Lyudmila A. Romanenko, Mikhail A. Pushilin, Pavel S. Dmitrenok and Tatyana A. Kuznetsova 1611 6-Bromo-5-hydroxyindolyl-3-glyoxylate from the Far Eastern Ascidian Syncarpa oviformis Elena A. Santalova, Vladimir A. Denisenko, Dmitry V. Berdyshev, Dmitry L. Aminin and Karen E. Sanamyan 1617 Analgesic Properties of New Pyrrolidinomorphinane Derivatives: Revealing Potential Pathways Ekaterina A. Morozova, Tatiana G. Tolstikova, Alexey V. Bolkunov, Margarita P. Dolgikh and Elvira E. Shul’ts 1621 2,4-Dihydroxypentanoic Acids: New Non-sugar Components of Bacterial Polysaccharides Alexander S. Shashkov, Nina A. Kocharova, Filip V. Toukach, Vadim V. Kachala and Yuriy A. Knirel 1625 Structural Analysis of Antibiotic INA 9301 from Amycolatopsis orientalis Alexander S. Shashkov, Dmitry E. Tsvetkov, Alexey A. Grachev, Olda A. Lapchinskaia, Maia F. Lavrova-Balashova, Valerii I. Ponomarenko, Genrikh S. Katrukha and Nikolay E. Nifantiev 1631 Structural Analysis of Fucoidans Maria I. Bilan and Anatolii I. Usov 1639 Guaiane Sesquiterpenoids from Jatropha curcas Xia-Chang Wang, Shi-Ping Ma, Jing-Han Liu and Li-Hong Hu 1649 Sesquiterpenes of the Brazilian Marine Red Alga Laurencia filiformis (Rhodophyta, Ceramiales) Bruno Lopes Antunes, Beatriz Grosso Fleury, MutueToyota Fujii and Valéria Laneuville Teixeira 1653 Inhibition of Mushroom Tyrosinase and Melanogenesis B16 Mouse Melanoma Cells by Components Isolated from Curcuma longa Jeong Ah Kim, Jong Keun Son, Hyun Wook Chang, Yurngdong Jahng, Youngsoo Kim, MinKyun Na and Seung Ho Lee 1655 Reaction of Furanoeremophilans with Pyridoxal Atsushi Torihata and Chiaki Kuroda 1659 A Bioactive Diterpene from Smallanthus sonchifolius Consolacion Y. Ragasa, Agnes B. Alimboyoguen, Sylvia Urban and Dennis D. Raga 1663 Antiproliferative Oleanane Saponins from Polyscias guilfoylei Giuseppina Cioffi, Antonio Vassallo, Laura Lepore, Fabio Venturella, Fabrizio Dal Piaz and Nunziatina De Tommasi `1667 Contents of Volume 3 (1-12) 2008 xiii Steroidal Saponins from the Rhizomes of Ruscus hypophyllum Yoshihiro Mimaki, Tsukasa Aoki, Maki Jitsuno, Akihito Yokosuka, Ceyda Sibel Kiliç and Maksut Coşkun 1671 A New Carbazole Alkaloid from Murraya koenigii Spreng (Rutaceae) Suvra Mandal, Anupam Nayak, Samir K. Banerjee, Julie Banerji and Avijit Banerji 1679 Secondary Metabolites from Teclea amanuensis (Rutaceae) from Tanzania Joseph J. Magadula, Modest C Kapingu, Zakaria H. Mbwambo and Dulcie A. Mulholland 1683 Chemical Constituents and Insecticidal Activity of Rollinia leptopetala (Annonaceae) Ângela M.C. Arriaga, Edinilza M.A. Feitosa, Telma L.G. Lemos, Gilvandete M.P. Santiago, Jefferson Q. Lima, Maria C.F. de Oliveira, Jackson N. e Vasconcelos, Francisco E. A. Rodrigues, Tathilene B. M. Gomes and Raimundo Braz-Filho 1687 New Pyrenocines from an Endophytic Fungus Karsten Krohn, Md. Hossain Sohrab, Siegfried Draeger and Barbara Schulz 1689 Chemoenzymatic Synthesis and Some Biological Properties of O-phosphoryl Derivatives of (E)-resveratrol Danilo Aleo, Venera Cardile, Rosa Chillemi, Giuseppe Granata and Sebastiano Sciuto 1693 Comparative Photobleaching Behavior of Hypocrellin A and Elsinochrome C Qian Zhao and Hong-Yu Zhang 1701 In vitro Cytotoxic Activity of Sesamin Isolated from Vismia baccifera var. dealbata Triana & Planch (Guttiferae) Collected from Venezuela Fabiola Salas, Janne Rojas, Antonio Morales, Maria E. Ramos-Nino and Nelida G. Colmenares 1705 A New Flavonoid Glycoside from the fern Dryopteris villarii Filippo Imperato 1709 Variation of Bioactive Secondary Metabolites in Hypericum triquetrifolium Turra from Wild Populations of Turkey Necdet Çamaş, Jolita Radušienė, Ali Kemal Ayan, Cüneyt Çırak, Valdimaras Janulis and Liudas Ivanauskas 1713 Anti-inflammatory Activity of Piper magnibaccum (Piperaceae) Emrizal, Farediah Ahmad, Hasnah M. Sirat, Fadzureena Jamaludin, Nik Musa’adah Mustapha, Rasadah M. Ali and Dayar Arbain 1719 Self-Organizing Maps as a New Tool for Classification of Plants at Lower Hierarchical Levels Mauro V. Correia, Marcus T. Scotti, Marcelo J. P. Ferreira, Sandra A. V. Alvarenga, Gilberto V. Rodrigues and Vicente P. Emerenciano 1723 Fatty Acid Composition of Heliotropium Species (Boraginaceae): A First Chemical Report on the New Species H. thermophilum Ahmet C. Gören, Gülendam Tümen, Ali Çelik and Simay Çıkrıkçı 1731 Comparative Analysis of the Essential Oils from two Asteraceous Plants Found in Nigeria, Acanthospermum hispidum and Tithonia diversifolia Adebayo A. Gbolade, Daniela M. Biondi and Giuseppe Ruberto 1735 Chemical Composition of the Essential Oil from Carramboa pittieri (Cuatrec.) Cuatrec. (Asteraceae) Luis B. Rojas, Rebeca Gutiérrez, Yndra Cordero de Rojas and Alfredo Usubillaga 1739 Antimicrobial Activity and Chemical Composition of Melaleuca genistifolia Leaf Essential Oil from the Northern Plains of India Ajai Kumar, Santosh K. Srivastava, Gaurav R. Dwivedi, Merajuddin Khan, Mahendra P. Darokar, Mohit Saxena, Kodakandla V. Syamsundar, Suaib Luqman and Suman P. S. Khanuja 1741 Number 11 New Drimane Sesquiterpenoids from Tidestromia oblongifolia Sandeep Chaudhary, Vladimir Thomas, Louis Todaro, Onica LeGendre, Stevan Pecic and Wayne W. Harding 1747 Three New Cassane Diterpenes from Caesalpinia pulcherrima Jun Cheng, Joy S. Roach, Stewart McLean, William F. Reynolds and Winston F. Tinto 1751 21,28-Epoxy-18β,21β-dihydroxbaccharan-3-one and Other Terpenoids from the Liverwort Lepidozia chordulifera T. Taylor Hildegard Zapp, Kerstin Orth, Josef Zapp, Joseph D. Connolly and Hans Becker 1755 A New Hyptadienic Acid Derivative from Hyptis verticillata (Jacq.)