University of Khartoum Graduate College Medical and Health Studies Board

Isolation and Structure Elucidation of Leishmanicidal Alkaloids from Argemone mexicana L. and Nauclea latifolia Smith.

By Omima Mekki Khider Mekki B. Sc. (1999), M. Sc. (2005) (U of K)

A thesis submitted in fulfillment of the requirement for the award of the degree of Doctor of Philosophy in Pharmaceutical Chemistry of University of Khartoum

Supervisor Prof. Sami Ahmed Khalid B. Pharm., M. Pharm., PhD.

2016 DEDICATION

This thesis is dedicated to my lovely family,

To my Parents for their lifelong devotion, patient and prayers

To my dearest Brothers and Sisters for their kind help and prayers

To my respectful supervisor prof. Sami for his kind support

Omima

TABLE OF CONTENTS

Contents Page No TABLE OF CONTENTS i ACKNOWLEDGMENTS v ABBREVIATIONS vii ENGLISH ABSTRACT ix ARABIC ABSTRACT xi LIST OF FIGURES xii LIST OF TABLES xiii 1. INTRODUCTION 1 1.1 Background information 1 1.2 Problem statement 2 1.3 Aim and Objectives 3 1. 4 Justifications 3 2. LITERATURE REVIEW 5 2.1 Neglected Tropical Diseases (NTDs) caused by protozoan 5 parasites 2.1.1 Contribution of Medicinal to NTDs 6 2.1.2 Ethnopharmacology of Sudanese medicinal plants with special 7 emphasis on NTDs 2.2 Leishmaniasis 8 2.2.1 Leishmanial chemotherapy 12 2.2.2 Medicinal plants reported to exhibited antileishmanial activity 13

i 2.2.3 Alkaloids from medicinal plants reported to exhibited 27 antileishmanial activity 2.3 Plants investigated in the present study 88 2.3.1 Argemone mexicana L. (Papaveraceae) 88 2.3.1.1 The botanical description of A. mexicana L. 88 2.3.1.2 Ethnopharmacological importance of A. mexicana L. 50 2.3.1.3 Phytochemistry of A. mexicana L. 53 2.3.1.4 Biosynthesis of benzophenanthridine alkaloids 58 2.3.1.5 Pharmacological activity of protoberberine and protopine 66 alkaloids 2.3.2 Nauclea latifolia Smith. (Rubiaceae) 63 2.3.2.1 The botanical description of N. latifolia Smith. 63 2.3.2.2 Ethnopharmacological importance of N. latifolia Smith. 63 2.3.2.3 Phytochemistry of N. latifolia Smith. 66 2.3.2.4 Biosynthesis of monoterpene indole alkaloid Strictosamide 44 66 2.3.2.5 Pharmacological activity of β-carboline alkaloids 68 3. MATERIALS AND METHODS 07 3.1 General experimental conditions 76 3.1.1 Organic solvents, glassware, chemicals and reagents 76 3.1.2 Spectroscopic techniques 76 3.1.3 Chromatography 71 3.1.4 Detection method on TLC by spraying reagents 72 3.2 Materials 72 3.2.1 Extraction and fractionation of plant materials 72

ii 3.3 Phytochemical screening 73 3.3.1 Isolation of compounds from Argemone mexicana L. 73 3.3.1.1 Isolation of compounds from petroleum ether fraction 73 3.3.1.2 Isolation of compounds from ethyl acetate fraction 74 3.3.2 Isolation of compounds from Nauclea latifolia Smith. 76 3.3.2.1 Isolation of compounds from petroleum ether fraction 76 3.3.2.2 Isolation of compounds from chloroform fraction 76 3.3.2.3 Isolation of compounds from ethyl acetate fraction 78 3.4 BIOLOGICAL ASSAYS 08 3.4.1 Materials 78 3.4.2 Antileishmanial assay 77 3.4.2.1 Maintenance of Parasites 79 3.4.2.1.1 Preparation of Biphasic medium (NNN medium) 79 3.4.2.1.2 Preparation of complete RPMI 1640 medium 79 3.4.2.1.3 Subculture of parasites 86 3.4.2.2 Preparation of sample 80 3.4.2.3 In vitro assay by colorimetric method 80 3.4.2.3.1 In vitro assay against extracellular promastigote 80 3.4.2.4 In vitro assay by macrophage method 81 3.4.2.4.1 Procedure for harvesting macrophages from the mice 81 3.4.2.4.2 In vitro assay against intracellular amastigotes 81

iii 3.4.3 Cytotoxicity assay 82 3.4.3.1 Cell-line 82 3.4.3.2 Method 82 4. RESULTS AND DISCUSSION 84 4.1 Secondary metabolites isolated from Argemone mexicana L. 84 4.1.1 Characterization of AM/1 as Sitost-4-en-3-one (β-Sitostenone) 84 113 4.1.2 Characterization of AM/2 as norchelerythrine 85 87 4.1.3 Characterization of AM/3 as berberine 10 87 4.2 Secondary metabolites isolated from Nauclea latifolia Smith. 71 4.2.1 Characterization of NL/1 as β-Sitosterol 114 71 4.2.2 Characterization of NL/2 as naucleficine 115 74 4.2.3 Characterization of NL/3 as strictosamide 44 78 4. 3Antileishmanial and cytotoxicity assays 162 4.4 Docking of the isolated compounds against leishmania enzymes 107 5. CONCLUSIONS AND RECOMMENDATIONS 110 5.1 Conclusions 11 0 5.2 Recommendations 11 2 6. REFERENCES 114

iv ACKNOWLEDGEMENTS

I would like to express my special appreciation and thanks to my supervisor Prof. Dr. Sami Ahmed Khalid. He has been a tremendous mentor for me. I would like to thank him for his precious and interesting suggestions, continuous support, patience, inspiration and immense knowledge. His guidance and encouragement helped me enormously in conducting my research and the writing up of this thesis.

I am deeply indebted to Prof. Dr. Atta ur Rahman of H.E.J, International Centre for Chemical and Biological Sciences (ICCBS), University of Karachi, Pakistan, for his comments and valuable suggestions. I would like to extend my thanks to Prof. Dr. Mohammed Igbal Choudhary for his kind guidance and his enjoyable lectures in NMR spectroscopy and other topics. I would like to thank both of them for providing me the access to the laboratory and research facilities. Without their precious support it wouldn’t have been possible to conduct significant part of this research.

Sincere thanks and appreciation to Prof. Dr. Souad Abd Elaziz for her prayers, continuous encouragement and follow up and for taking over all my responsibilities at the University of Science and Technology during conducting my research in Pakistan. I am so grateful and obligated to the ex- dean of the Graduate College, University of Khartoum, Prof. Dr. Mohammed Ahmed Abu Alnour for his support and understanding of all circumstances and health conditions I went through during this research.

v My sincere thanks are due to Mr. Fouad Abd Aljalil, Faculty of Pharmacy, University of Khartoum, for his technical assistance in the preparation of the extracts.

I am deeply indebted to Dr. Farzana Navid for her kind support and being a caring and loving sister as well as a genuine friend. Special thanks to my fellow lab mates at H.E.J, ICCBS, for the stimulating discussions and the great fun we have had during our works. In particular, I am so grateful to the PhD fellow Mujeeb ur Rahman, Dr. Shabbir Husein, PhD fellow Ismaeel and Dr. Adighari. Thanks also to all technical staff of NMR, MS, HPLC laboratories and IT staff at ICCBS for their unlimited technical support during my stay in ICCBS.

I would like to extend my thanks to Dr. Sammer Yousuf, Miss. Samreen Khan and Miss. Nida Ghouri for performing the antileishmanial assay. I am also obliged to Dr. Omer Mohammed Elhaj for performing the cytotoxicity assays.

I do sincerely appreciate the support of Dr. Asaad Khalid and Dr. Mohammed Ahmed Mesaik for facilitating my mission at the ICCBS.

I would like to express my sincere thanks and gratitude to Prof. Dr. Moawia Mukhtar, and his lab. team, Institute of Endemic Diseases, University of Khartoum, for hosting me, providing me the access to the laboratory, and teaching me how to culture the leishmania parasite and macrophage cells.

Great thanks and appreciation to University of Khartoum for offering me the scholarship and the opportunity to achieve my post graduate studies.

vi ABBREVIATIONS

NTDs: Neglected Tropical Diseases CL: Cutaneous leishmaniasis MCL: Mucocutaneous leishmaniasis VL: Visceral leishmaniasis TDR: The special program for research and training in tropical diseases WHO: World health organization GDP: Gross domestic product HIV: Human immunodeficiency virus HAART: Highly active antiretroviral therapy DNDi: Drug for neglected diseases initiative DCM: Dichloromethane Pro: Promastigote Ams: Amastigote N. R.: Not reported DOPA: 3,4-dihydroxyphenylalanine TDC: Tryptophan decarboxylase G10H: Geraniol 10 hydroxylase SLS: Secologanin synthase STR: Strictosidine synthase SAP: Secreted aspartic protease MTT: 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (dye) DMEM: Dulbecco's Modified Eagle's Medium

vii MEM: Minimum Essential Medium eagle DMSO: Dimethyl sulfoxide EDTA: Ethylenediaminetetraacetic acid HEPES: 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (buffer) RPMI 1640: Roswell Park Memorial Institute medium PBS: Phosphate buffered saline FBS: Fetal bovine serum FCS: Fetal calf serum NNN medium: Novy-MacNeal-Nicolle medium rpm: Revolutions per minute PI: Percentage inhibition

IC50: Inhibition concentration SD: Standard deviation

viii ABSTRACT

Introduction: Leishmaniasis a group of clinical diseases affecting millions of the world populations in 88 countries. Accordingly, this disease is considered as one of the Neglected Tropical Diseases (NTDs) besides malaria, sleeping sickness, mycetoma and other fifteen NTDs. Chemotherapy for leishmaniasis is still deficient and there is an urgent need to discover novel antileishmanial agents, hence most of the currently available drugs have serious limitations such as long-term administration, unaffordable cost, toxicity, and developing resistance by these parasites.

The objectives of this study is to isolate and elucidate the chemical structures of new anti-leishmanial secondary metabolites against both the promastigotes and amastigotes stages with special reference to alkaloids occurring in Argemone mexicana L. and Nauclea latifolia Smith, which are commonly growing plants in Sudan.

Methods: The extraction of plant materials was followed by bioactivity- guided fractionation using the promastigotes of L. donovani, L. major and L. tropica of the crude ethanolic extracts of A. mexicana and N. latifolia and their respective organic fractions as well as the water residue by colorimetric method. Isolation of pure compounds was achieved by high-performance liquid chromatography (HPLC). The isolated compounds were subjected to one- and two-dimensional Nuclear Magnetic Resonance (NMR) with special emphasis on 1D-NMR (1H, 13C, DEPT), 2D-NMR (COSY, HSQC, HMBC, NOESY) as well as UV, and mass spectrometry.

Results: Bioactivity-guided fractionation revealed a remarkable difference in susceptibility of the three leishmania species. The crude ethanolic extract of A. mexicana exhibited the most prominent activity against the promastigotes of L. donovani (IC50 11.39 μg/mL) followed by the water residue (23.93 μg/mL) which is indicative of the hydrophilicity of the bioactive compounds.

ix The crude ethanolic extract and water residue of Nauclea latifolia exhibited weak activity against the promastigotes of L. donovani with IC50 of 46.86 and 30.45 μg/mL, respectively.

Among the purified isolated alkaloids of Nauclea latifolia, strictosamide inhibited the proamastigotes of both L. major and L. tropica with IC50 of 77.67±1.11 and 80.40±3.00 μg/mL, respectively. The structurally related β- carboline alkaloid, naucleficine, however, showed no inhibition. Similarly, the isoquinoline alkaloid, norchelerythrine, isolated from A. mexicana, exhibited no leishmanicidal activity against L. major and L. tropica.

Attempting computational chemistry by docking of the isolated compounds against 14 leishmanicidal targets revealed that strictosamide has a relatively strong binding to certain protein targets such as phosphodiesterase B1 (LmajPDEB1) (PDB: 2R8Q) and Uridine 5'-monophosphate synthase (LdUMPS) (PDB: 3QW4) among all the isolated alkaloids.

Conclusion: The aerial part of Argemone mexicana yielded two benzophenanthridine alkaloids, norchelerythrine and berberine besides the tetracyclic terpenes, sitost-4-en-3-one (β-sitostenone. The extract of the root bark of Nauclea latifolia yielded the two β-carboline alkaloids, naucleficine and strictosamide besides the ubiquitous β-sitosterol.

Further research is required to reveal the possible mechanism of action of the isolated compounds.

x خالصة البحث

المقدمة: الليشمانيا هي مجموعة امراض سريرية تهدد ماليين من سكان العالم في حوالي 88 قطر. لذلك يعتبر هذا المرض من االمراض االستوائية المهملة بجانب المالريا، مرض النوم، المايسوتوما وغيرها. االدوية المستخدمة لعالج الليشمانيا لها عدة اثار جانبية كفترة العالج الطويلة، غالء االدوية ، السمية وكذلك مقاومتها من قبل الطفيل، مما يدعو للحاجة الماسة الكتشاف عقار جديد.

الهدف من هذه الدراسة عزل وتحديد البنية الكيميائية لمضادات ليشمانيا جديدة ضد الطور السوطي الخرجي والطور الخلوي الداخلي للطفيل. مع التركيز بشكل خاص علي القلويدات الموجودة في نباتي االرجيمون والكرمدودة التي تنمو في السودان.

الطريقة: استخالص اجزاء النبات متبوعة بفحص النشاط الحيوي للمستخلصات ضد الطور الخارجي السوطي لالنواع الثالثة من الليشمانيا للمستخلصات االيثانولية للخام النباتي لالرجيمون والكرمدودة واجزاء كل منها في االيثر البترولي ، الكلوروفورم، خالت االيثيل والماء بالطريقة اللونية. استخدمت تقنيتي كروماتوغرافيا العمود وكروماتوغرافيا الطبقة الرقيقة التحضيرية عالية االداء في عزل المركبات النشطة حيويا. تم توضيح هيكل المركبات المعزولة باستخدام طرق طيفية حديثة مختلفة: الرنين المغناطيسي، األشعة فوق البنفسجية ومطياف الكتلة.

النتائج: دلت نتائج فحص النشاط الحيوي علي فروقات ملحوظة في استجابة االنواع الثالثة من الليشمانيا ضد المستخلصات. اظهر المستخلص االيثانولي الخام والمائي لنبات االرجيمون النشاط االبرز ضد الطورالسوطي الخارجي للشمانيا donovani وقد كان التركيز المثبط )%56( يعادل 11.37 ميكروجرام/مل و23.73 ميكروجرام/مل علي التوالي. وهذا يدل علي ان المركبات النشطة بيولوجيا تركزت في المحلول المائي. كما اظهر المستخلص االيثانولي الخام والمائي لنبات الكرمدودة نشاطا ضعيفا ضد الطور السوطي الخارجي للشمانيا donovani وكانت التراكيز المثبطة ) %56( 86.86 و 36.85 ميكروجرام علي التوالي.

من بين القلويدات المعزولة نقية من الكرمدودة، strictosamide وقد قام بتثبيط الطور السوطي الخارجي لكل من اللشمانيا major وtropica وكانت قيم التراكيز المثبطة 77.67 و 86.86 ميكروجرام/مل، على التوالى. غير ان القلويد بيتا- كاربولين ، naucleficine، لم يظهر أى نشاط مضاد للشمانيا. وبالمثل فان قلويد آيسوكينولين، norchelerythrine المعزول من نبات االرجيمون، لم يظهر أى نشاط ضد الليشمانيا major و tropic.

أظهرت محاولة االلتحام الجزيئى للمركبات المعزولة ضد أربعة عشر من انزيمات الليشمانيا المستهدفة عن وجود عالقة ربط قوية ل strictosamide تجاه phosphodiesterase B و Uridine 5'-monophosphate synthase من بين كل القلويدات المعزولة.

الخالصة: تم بنجاح عزل اربعة قلويدات واثنان من التربينات رباعية الحلقة وقد تم تحديد البنية التركيبية باستخدام طرق طيفية حديثة. المركبات المعزولة من نبات االرجيمون كانت قلويدات ايسوكينولية، berberine,norchelerythrine باالضافة الي التربين رباعي الحلقة β-sitostenone . بينما مستخلص قلف الجذور لنبات الكرمدودة اعطي اثنان من القلويدات بيتاكاربولين، naucleficine و strictosamide باالضافة الي التربين رباعي الحلقة β-sitosterol.

المزيد من البحث لتجربة المركبات المعزولة داخل الخلية لمعرفة ميكانيكية عملها.

xi LIST OF FIGURES

Title of figure Page No Figure 1. Life cycle of leishmania parasite 16

Figure 2. Chemical structures of reported antileishmanial alkaloids 86

Figure 3. The main isoquinoline alkaloids of Argemone mexicana 58

Figure 4. Biosynthesis of (+)-Reticuline 92, a precursor of several 56 isoquinoline alkaloids

Figure 5. Formation of 9,10- or 10,11- substituted intermediates from 57 (+)-Reticuline 92 in the biosynthesis of protoberberine alkaloids

Figure 6. Biosynthesis of the benzophenanthridine alkaloids 57 Chelidonine 96 and Sanguinarine 88 from the common precursor (+)- Reticuline 92

Figure 7. Biosynthesis pathway of monoterpene indole alkaloid 67 Strictosamide 44

Figure 8. Chemical structures of indole alkaloids from Nauclea sp that 67 exhibited many biological activities

Figure 9. Selected COSY and HMBC Correlations of naucleficine 115 76

Figure 10. Selected HMBC Correlations of strictosamide 44 160

Figure 11. Interactions of β-Sitosterol and LmajHSP90. 109

Figure 12. Interactions of β-Sitosterol and LmajPDEB1. 109

xii LIST OF TABLES

Title of table Page No Table 1. Plants reported as antileishmanial 18

Table 2. Reported antileishmanial alkaloids 28

Table 3.1HNMR and 13CNMR spectral data for compound AM/1113 86

Table 4.1HNMR and 13CNMR spectral data for compound AM/2 85 88

Table 5.1HNMR and 13CNMR spectral data for compound AM/3 10 70

Table 6. 1HNMR and 13CNMR spectral data for compound NL/1 114 73

Table 7.1HNMR and 13CNMR spectral data for compound NL/2 115 77

Table 8. 1HNMR and 13CNMR spectral data for compound NL/3 44 161

Table 9. IC50 of the two plants extracts/fractions against the 163 promastigotes of L. donovani, L.major and L.tropica compared with the IC50 of the standard drugs Amphotericin B and Pentamidine by in vitro method (colorimetric)

Table 10. IC50 of the extracts/fractions against the amastigotes of L. 166 major and L. tropica compared with the IC50 of the standard drug, sodium stibogloconate (Pentostam®) (13.47±0.17 μg/mL) by macrophages method.

Table 11. IC50 of the pure compounds isolated from the two plants 167 against Leishmania major and Leishmania tropica compared with the IC50 of the standard drugs Amphotericin B (0.29±0.05 μg/mL) and Pentamidine (5.09±0.09 μg/mL) by in vitro method (colorimetric)

Table 12. Docking Binding free energies of the top ranked compounds 168 with leishmania enzymes

xiii 1. INTRODUCTION

1.1 Background information Protozoan parasites are among the most common chronic infections that occur primarily in rural and poor urban areas of tropical and subtropical regions around the world. They are responsible for a large number of severe and widespread diseases including malaria, leishmaniasis, Chagas’ disease (American trypanosomiasis), and sleeping sickness (African trypanosomiasis). These diseases belong to the group of Neglected Tropical Diseases (NTDs), since they are strongly linked with poverty and there is a lack of commercial markets for potential drugs. These diseases affect individuals throughout their lives, causing a high degree of morbidity and physical disability and, in certain cases, gross disfigurement. Patients can face social stigmatization and abuse as a result of contracting these diseases. The disability and the poverty associated with these diseases constitute large burdens on the health and economic development of low-income and middle-income countries in Africa, Asia, and the Americas. Overall global prevalence is approximately 550 million cases, and close to 2.7 billion people living in endemic areas are at risk of contracting any of these diseases. In total, there are about 280 million new cases each year causing important health and socio-economic problems where these diseases are endemic. Chemotherapy remains one of the key measures used to control the intolerable burden of protozoan parasitic and other tropical diseases, but most of the available drugs are no longer effective due to drug resistance. Moreover, some of those that are still effective suffer from problems associated with toxicity, compliance and high cost, resulting in an urgent need for new drugs (Osorio et al., 2008).

1

1.2 Problem statement Leishmaniasis a group of clinical diseases suffered by millions around the world, and affecting 88 countries in Africa, Asia, Europe and America, 72 of which are developing countries and 13 are among the least developed. The spectrum of the diseases divided into three major syndromes: cutaneous leishmaniasis (CL), mucocutaneous leishmaniasis (MCL) and visceral leishmaniasis (VL). Annual incidences estimated at 1.5 million cases of the cutaneous forms (CL and MCL) and 500,000 cases of the visceral form (VL), resulting in approximately 51,000 deaths per year. Overall prevalence is 12 million people and the population at risk is 350 million. However, this estimated global burden of disease is believed to be inaccurate partly due to the passive case detection data used to estimate the disease prevalence in many endemic countries. Apparently, for each symptomatic case, there are estimated to be 10 asymptomatic infections. The total burden of Disability Adjusted Life Years (DALY) is 2.09 million, with 840,000 for women and 1.25 million for men. The Special Program for Research and Training in Tropical Diseases (TDR) has classified leishmaniasis as a group of emerging or uncontrolled diseases (Osorio et al., 2008).

Leishmaniasis is produced by at least 20 species of the protozoan Leishmania. Chemotherapy for leishmaniasis is still deficient. Most of the drugs have one or more limitations such as long-term administration, the route of administration is non orally, unaffordable cost and toxicity or even worse, inefficacy due to development of resistance in the parasite. Although these drugs constitute the main antileishmanial chemotherapy and have been used for over 50 years, information about their chemistry or precise mode of action and the identity of the biologically active components are uncertain (Osorio et

2 al., 2008). Therefore, there is an urgent need for new medicine , which are safe, efficient, effective, cheap, easy to administer, and for new lead compounds with novel mechanisms of action.

1.3 Aim and objectives The main aim of the present work is to investigate the anti-leishmanial activity of the secondary metabolites of two plants grown in Sudan, namely the aerial parts of Argemone mexicana L. and the root bark of Nauclea latifolia Smith. The specific objectives are: 1. To subject the plant extracts to bioactivity- guided fractionation to isolate the bioactive leishmanicidal compounds with special interest on alkaloids. 2. To elucidate the structures of the isolated compounds using modern spectroscopic methods, 3. To assess the anti-leishmanial activity against both the promastigotes and amastigotes.

3

1.4 Justifications The search for new and effective agents against leishmania and other diseases has led to increase interest in existing information about the remedies of these diseases from natural sources principally the plants. Efforts have been devoted and directed by many researchers to the isolation and characterization of the biological active principle of the plants. Natural products literature provides a growing research on plant derived antileishmanial agents and several natural products so far have been discovered with excellent activity against leishmania parasites. Alkaloids are the most potent therapeutic compounds of natural origin, they are an important class of secondary metabolites that occur in plants and also in certain higher animals and marine invertebrates (Okwu and Uchenna, 2009; Mishra et al., 2011). Alkaloids containing plants and their biosynthesized alkaloids have a remarkable potential to provide pharmaceutical and biological agents contributing to the development of future antiparasitic drugs (Osorio et al., 2008). It has been reported that alkaloids exhibiting significant anti-leishmanial activities (Mishra et al., 2011). Khalid (2012) reviewed the bioactivity of some isolated antiparasitic compounds from Sudanese medicinal plants against NTDs and still several compounds waiting to be discovered.

4

2. LITERATURE REVIEW

2.1 Neglected Tropical Diseases (NTDs) caused by protozoan parasites NTDs are a group of chronic disabling infections affecting more than a billion people worldwide, mainly in Africa and mostly those living in remote rural areas, urban slums or conflict zones. People suffering from NTDs are predominantly afflicted by poverty and they constitute an unattractive market to private-sector research and development (R&D) investment. These diseases not only affect our health but also represent a vicious cycle of socioeconomic events which reinforce and feedback on each other, leading towards inescapable poverty of a sizable number of the population (Khalid, 2012).

The global threat posed by NTDs compelled the World Health Organization (WHO) to release its first report on the situation five years ago (WHO, 2010). The number of NTDs enumerated in this report reached 17, including three soil-transmitted helminthiases. Although malaria is not mentioned among the NTDs in this report, possibly due to a resurgence at the global level of increased funding by various international bodies (e.g. Roll Back Malaria Initiative, RBM) and philanthropic organizations (e.g. The Bill & Melinda Gates Foundation) during the last decade, malaria still remains one of the NTDs; this is mainly due to the fact that the death toll from this disease in Africa constitutes over 90% of the total number of global death and due to its direct/indirect economic impact which results in an estimated US$ 12 billion annual income loss in sub-Saharan Africa, which translates to a 1.3% annual loss in gross domestic product (GDP) in this malaria endemic African region (Berger et al., 2010). Recent publications still include malaria together with

5 other NTDs such as leishmaniasis, trypanosomiasis, schistosomiasis, onchocerciasis, lymphatic filariasis and dengue fever (Chatelaine and Loset, 2011). Each year, there are between 300 and 500 million clinical cases of malaria; estimates of the number who die from the disease range between 1.0 and 2.5 million annually. A disproportionate number of deaths from malaria occur among the poor, and about half of those who die are children and pregnant women. Currently, there is no vaccine for malaria, and the parasite has already developed resistance against almost all the currently available antimalarials. Unfortunately, new reports indicate the emergence of resistance against artemisinin derivatives as well (Khalid, 2012).

2.1.1 Contribution of medicinal plants to NTDs The fascination of natural products, especially from plants with known medicinal properties, goes back to ancient times. The utilization of plants for and as medicines is old as civilization. Different traditional medicinal systems have been for a long time, making use of plants as effective medicines to treat many harmful diseases (Jyothirmayi and Prasad, 2012). At the beginning of the 19th century the discovery of pure compounds as active constituents from plants were described, and the art of exploiting natural products has become part of the molecular sciences (Kayser et al., 2002). Impressively within the last 25 years a large fraction of drugs recently approved have their origin in nature. Even more all approved anti-tumor drugs since the 1940s, of which more than 50% are of natural origin. However, many drugs used against infectious diseases are natural products or derivatives. Many reports estimated that approximately 80% of the population in developing countries still relies on traditional medicine for their primarily health care (Karou et al., 2011). About one quarter of the present prescription drugs dispensed by community

6 pharmacies in the United States contain at least one active principle originally derived from plant materials (Agyare et al., 2006). A milestone in the history of a remedy used successfully since ancient days against a protozoan infection would be the natural alkaloid quinine from Cinchona succiruba (Rubiaceae) and its subsequent development as drug which is still used successfully and increasingly against malaria today. Discovering untapped natural sources of novel antiprotozoal compounds from nature remains a major challenge and a source of novelty in the area of combinatorial chemistry and genomics (Schmidt et al, 2012a; Kayser et al., 2002). There are more than 2,70,000 higher plants existing in this planet, but so far less than 10% of recorded flora has been explored phytochemically as well as clinical evaluation for various biological activities (Reddy, 2010).

2.1.2 Ethnopharmacology of Sudanese medicinal plants with special emphasis on NTDs Sudan as an African tropical country is confronted with many parasitic and infectious diseases prevailing on this continent. Similar to other developing countries, traditional medical practices play an important role in Sudan. Herbal drugs are of major importance in Sudanese folk medicine. This was documented during comprehensive ethnobotanical investigations (El Kamali and Khalid, 1996; El Ghazali et al., 1994, 1997; El Kamali and El Khalifa, 1997, 1999; El Tahir et al., 1998, 1999a, 1999b; Ali et al., 2002; Khalid et al., 2012; Khalid, 2012).

A number of systematic attempts have been made to verify the claimed antiparasitic uses of Sudanese plants and to detect and/or isolate their bioactive agents (Khalid et al., 1986, 1989, 1998; El Kamali and El Khalifa, 1997; El

7

Tahir et al., 1998, 1999a,b). Among other natural products, the limonoid 2,6- dihydroxyfissinolide (Khalid et al., 1998) from Khaya senegalensis and the tetranortriterpenoid gedunin from Azadirachta indica (Khalid et al., 1989) were found to exhibit good antimalarial activity. The bioactivity of some isolated bioactive compounds from Sudanese medicinal plants against NTDs has been reviewed (Khalid, 2012). Inhibitory effects of Sudanese plants on HIV-1 replication and HIV-1 protease were also investigated (Hussein et al., 1999; Ali et al., 2002), as well as their declared antibacterial activities (Elegami et al., 2001). Ali et al., 2002 verified the ethnopharmacological uses of some selected Sudanese plants commonly employed to treat malaria and similar tropical diseases. The ethnobotanical information thus served as basis for the selection of the plants studied. They investigated the activities of diverse plant extracts against Plasmodium falciparum, Trypanosoma spp. and Leishmania donovani, their inhibitory activity towards HIV-1 reverse transcriptase and p56lck tyrosine kinase was also assessed.

2.2 Leishmaniasis Leishmaniasis represents a set of severe human diseases. These diseases are endemic in Africa, America, Indian sub-continent and in sub-tropical southwest Asia as well as the Mediterranean. Leishmania species are obligatory intracellular protozoan parasites exist in macrophages of their mammalian hosts. The extracellular stage, the promastigote, is transmitted into subcutaneous tissue in the human host (or animal) during the bite of an infected sandfly vector belong to the Phlebbotomus (Old World) and Lutzomyia (New World), both subfamily Phlebotominae, family Psychodidae. The promastigote is phagocytosed by a mononuclear phagocyte, after which it

8 converts into the obligatory intracellular form, the amastigote (Schmidt et al., 2012a).

The leishmania promastigotes are transmitted by sandfly to vertebrate hosts e.g. canines, marsupials, edentates and rodents. Once inside the blood stream of reservoirs for the disease, promastigotes are phagocytosed by the mononuclear phagocytic cells and are transformed to amastigotes that multiply by means of binary fission. On lyse of host cell, the free parasites spread to new cells and tissues of different organs including the spleen, liver and bone marrow. Amastigotes in the blood as well as in the monocytes are ingested during a blood meal by female sandfly. Once ingested, the amastigotes migrate to the midgut of the sandfly and transform into the promastigotes. After a period of four to five days, promastigotes move forward to the oesophagus reach to salivary glands of the sandfly. Infected sandfly during the second blood meal regurgitates the infectious promastigotes from its pharynx into the blood stream of the host vertebrates and the life cycle is repeated (Mishra et al., 2011).

9

Figure 1. Life cycle of leishmania parasite

Leishmania parasites were independently described by William Leishman and Charles Donovan in 1903 (Ross, 1903), but were previously observed by David D. Cuningham in 1885 and Peter Borovsky in 1989. The genus leishmania was proposed by James Wright in 1903 (Monzote, 2009).

There are about 20 known species of leishmania that have been associated with various disease forms in man. The severity of the disease ranges from the disfiguring cutaneous type in which nodules form at the site of infection, leaving scars upon healing, to the lethal visceral leishmaniasis. Some forms of the disease are anthroponotic (transmitted between humans), while others are zoonotic (involving an animal reservoir). Leishmaniasis is classified into three groups: cutaneous leishmaniasis (CL), mucocutaneous leishmaniasis (MCL) and visceral leishmaniasis (VL) on the basis of their clinical symptoms. There

10 are two general forms, visceral leishmaniasis (VL) caused by two species of leishmania, Leishmania donovani and Leishmania infantum (chagasi) and tegumentary leishmaniasis (TL), as cutaneous leishmaniasis (CL) and disfiguring mucocutaneous leishmaniasis (MCL). TL caused by several dermatropic species of leishmania eg.; L. major, L. tropica, L. aethiopica, L. braziliensis, L. panamensis, L. amazonensis and L. mexicana. Leishmaniasis affects around 10 million people worldwide, with an annual incidence of approximately two million new cases, 350 million are living at the risk to be infected, and causes approximately 50,000 death cases annually. VL causes 500,000 new cases annually globally, 90% of the new cases occurring in just five countries, India, Bangladesh, Brazil, Nepal and Sudan. Tegumentary forms of the disease affects 1,500,000 people. Although not a killing disease as is VL, disfigurement, disability and social and psychological stigmas are all severe consequences of TL (Schmidt et al., 2012a; Osorio et al., 2008).

The leishmanial infection development depends on the parasite properties, such as infectivity, pathogenicity and virulence, beside the heterogeneity of the host regulatory response resulting in different clinical manifestations. There are a rising number of reports of leishmania/human immunodeficiency virus (HIV) co-infections across the world. leishmania/HIV co-infections has been globally controlled in Southern Europe since 1997 by highly active anti retroviral therapy (HAART), but it appears to be an increasing problem in other countries such as Ethiopia, Sudan, Brazil or India where both infections are becoming more and more prevalent. The situation is particularly alarming in southern Europe, where 50-75% of adult VL cases are HIV positive and among the 45 million people infected by HIV worldwide, an estimated one- third lives in the zones of endemic leishmania infections. To date, the greatest

11 prevalence of leishmania/HIV co-infections has been in the Mediterranean basin. Among more than 2.000 cases notified to the WHO, 90% of them belong to Spain, Italy, France and Portugal (Mishra et al., 2011).

2.2.1 Leishmanial chemotherapy The drugs recommended for the treatment of leishmaniasis are pentavalent antimonials (SbV compounds: sodium stibogloconate, Pentostam® and meglumine antimoniate, Glucantime®), these drugs since 1950s are problematic due to toxicity/side effects, and teratogenicity, as well as increasing resistance. Pentamidine, Pentacarinat® is used as alternative drug in cases resistant to antimonials, but severe side effects arising in the required long term treatment are reported (Schmidt et al., 2012a).

Amphotericin B (AmB), the antifungal polyene antibiotic is used as a second- line antileishmanial drug in developing countries and as a first-line drug in the industrialized countries. Miltefosine, Impavido®, the oral antineoplastic agent, an alkylphosphocholine, is available on the market for the treatment of leishmaniasis since 2002 (Schmidt et al., 2012a). In a combination with SbV compounds, interferon-γ can be administrated especially in immunosuppressed patients; it stimulates the phagocytic activity of macrophages in order to kill amastigotes that reside in them. However this treatment is cost intensive and due to lack of clinical trials it remains limited today (Schmidt et al., 2012a).

12

The triazole, the antimycotic posaconazole, currently under investigation by the Drug for Neglected Diseases initiative (DNDi) against Chagas disease, was found to be active against Leishmania amasonensis and Leishmania donovani in rodent models. Oxaboroles and nitroimidazole are compounds that investigated further by the DNDi showed in vivo efficacy and are currently in pharmacokinetic trials (Schmidt et al., 2012a).

Tafenoquine, the antimalarial compound was reported to be highly active in vitro and in a rodent model against leishmania. The orally administrated antimalarial, artemisinin was effectively reduced the parasite burden in BALB/c model of VL (Schmidt et al., 2012a). Although the causative agents of leishmaniasis have been known and studied since 1903 (Ross, 1903), to date an effective cure for the disease does not exist.

2.2.2 Medicinal plants reported to exhibited antileishmanial activity A bewildering array of secondary metabolites has been reported to exhibit remarkable antileishmanial activity. Table 1 compiles those plants reported to have antileishmanial activity.

13

Table 1. Plants reported as antileishmanial Botanical source Extract/Fraction Bioactivity/life stage Leishmania spp References Acanthaceae N. R. In vitro (Pro.) L. donovani Carvalho and Ferreira, Acanthus illicifolius (Leaves) 2001 Andrographis paniculata (Roots) N. R. N. R. L. infantum Schmidt et al., 2012a Agavaceae Ethanol N. R. L. amazonensis Rocha et al., 2005 Yucca filamentosa L. (c) Anacardiaceae N. R. In vitro (axenicAms.) L. donovani Schmidt et al., 2012a Campanosperma panamense (N. R.) Annonaceae N. R. In vitro & in vivo L. amazonensis Rocha et al., 2005 Annona haematantha Miq (N. R.) (N. R.) Annona glauca Thonn (Seeds) DCM In vitro (Pro.) L. braziliensis. Carvalho and Ferreira, L. donovani. 2001 L. amazonensis Schmidt et al., 2012a Annona muricata L (Pericarp) Ethyl acetate N. R. L. braziliensis Rocha et al., 2005 L. panamensis Annona muricata (Seeds) N. R. In vitro (Pro.&Ams.) L. chagasi Schmidt et al., 2012a Annona senegalensis (Seeds) DCM N. R. L. donovani Schmidt et al., 2012a L. major Annona senegalensis (Seeds) Methanol & hexane In vitro (Pro.). L. major Schmidt et al., 2012a Annona aff. Spraguei Saff (Seeds) Chloroform N. R. L. braziliensis Rocha et al., 2005 L. panamensis L.infantum Annona squamosa (Leaves) N. R. In vitro (Pro.&Ams.) L. chagasi Schmidt et al., 2012a Duguetia furfuracea (Stem bark) N. R. In vitro (Pro.) L. braziliensis Schmidt et al., 2012a Friesodielsia obovata (N. R.) N. R. In vitro (axenicAms.) L. donovani Schmidt et al., 2012a Guatteria amplifolia (N. R.) N. R. N. R. L. panamensis Schmidt et al., 2012a L. mexicana Guatteria foliosa Benth (Stem bark) Alkaloid fraction N. R. L. amazonensis Rocha et al., 2005 L. braziliensis L. donovani Key: c: Data incomplete derived from an abstract; Pro.: promastigote; Ams.: amastigotes; DCM: dicloromethane; N. R.: not reported

14

Table 1. Plants reported as antileishmanial (Continued…) Botanical source Extract/Fraction Bioactivity/life stage Leishmania spp References Annonaceae Alkaloid fraction. N. R. L. amazonensis Rocha et al., 2005 Cardiopetalum calophyllum Schldl L. braziliensis (Leaves + Stem) L. donovani Duguetia spixiana Mart (Stem bark) Guatteria schoburgkiana Mart. (Leaves + stem bark) Oxandra espintana (Spruce) Baillon (Leaves + Stem bark)

Xylopia aromatica (Lam.) Mart (Leaves + Stem bark) Polyalthia longifolia var. pendula (N. R.) N. R In vitro & in vivo L. donovani Schmidt et al., 2012b (Ams.) Polyalthia macropoda king (Stem bark) N. R. In vitro (Pro.) L. donovani Carvalho and Ferreira, 2001 Rollinia emarginata Schdl. (Stem bark) DCM fraction of In vitro (Pro.) L. amazonensis Schmidt et al., 2012a methanol extract L. braziliensis L. donovaniz Unonopsis buchtienii R. E. Fries (N. R.) N. R. In vitro (Pro.) L. donovani Rocha et al., 2005 L. major Carvalho and Ferreira, 2001 Apiaceae N. R. In vitro (Pro.) L. major Schmidt et al., 2012a Ferula szowitsiana (N. R.) Apocynaceae N. R. In vitro (axenic Ams.) L. donovani Schmidt et al., 2012a Aspidosperma vargasii (Bark) Alstonia scholaris R.Br. (Stem) Ethanol N. R. L. donovani Rocha et al., 2005 Holarrhena curtisii King & Gamble Ethanol N. R. L. donovani Rocha et al., 2005 (Leaves) Mandevilla antennacea K. Schum. Ethanol N. R. L. amazonensis Rocha et al., 2005 (Leaves + stem) L. braziliensis Pentalinon andrieuxii (N. R.) N. R. N. R. Leishmania sp. Schmidt et al., 2012b Peschiera australis Miers. (Stem) Chloroform fraction In vitro (Pro.) & L. amazonensis Delorenzi et al., 2001 of Ethanol extract in vivo (Ams.) Key: c: Data incomplete derived from an abstract; Pro.: promastigote; Ams.: amastigotes; DCM: dicloromethane; N. R.: not reported

15

Table 1. Plants reported as antileishmanial (Continued…) Botanical source Extract/Fraction Bioactivity/life stage Leishmania spp References Apocynaceae Ethanol In vitro & in vivo L. amazonensis Rocha et al., 2005 Peschiera van. heurkii (Muell. Arg.) L. (Ams.) L. braziliensis Carvalho and Ferreira, Alloorge (Leaves + stem bark) 2001 Picralima nitida Th. &H. Dur. (Seeds) Chloroform N. R. L. donovani Rocha et al., 2005 Tabernaemontana obliqua (Miers) Leen Methanol N. R. L. amazonensis Rocha et al., 2005 wenb (Leaves) Araliaceae Ethanol In vitro (Pro.&Ams.) L. amazonensis Rocha et al., 2005 Hedera helix L. (Leaves) L. infantum Carvalho and Ferreira, L. tropica 2001 Cussonia zimmermannii (Root bark) Petroleum ether In vitro (axenic Ams.) L. donovani Schmidt et al., 2012a Oreopanax species (Leaves) Ethanol N. R. L. braziliensis Rocha et al., 2005 Asclepiadaceae Methanol N. R. L. major Rocha et al., 2005 Periploca graeca L. (twig) Gongronema latifolia Benth (Leaves) Methanol N. R. L. donovani Rocha et al.,2005 N. R. In vitro (Pro.) L. major Schmidt et al., 2012a Asparagus africanus (Roots) Ethanol N. R. L. amazonensis Rocha et al., 2005 Acanthospermum hispidum DC. L. braziliensis (Entire plant) Achyrocline flaccida (Weinm.) DC. Ethanol N. R. L. braziliensis Rocha et al., 2005 (Entire plant) Ageratina pentlandiana (DC.) K. & R. Ethanol N. R. L. braziliensis Rocha et al., 2005 (Leaves) L. amazonensis Ageratum conyzoides (N. R.) N. R. N. R. L. donovani Schmidt et al., 2012a Artemisia herba-alba Asso. (c) Aqueus In vitro (N. R.) L. tropica Rocha et al., 2005 Baccharis retusa (N. R.) N. R. In vitro (Pro.) L. braziliensis Schmidt et al., 2012a L. amazonensis In vitro (Ams.) L. major L. chagasi Key: c: Data incomplete derived from an abstract; Pro.: promastigote; Ams.: amastigotes; DCM: dicloromethane; N. R.: not reported

16

Table 1. Plants reported as antileishmanial (Continued…) Botanical source Extract/Fraction Bioactivity/life stage Leishmania spp References Asteraceae Ethyl acetate N. R. L. braziliensis Rocha et al., 2005 Baccharis salicifolia (R. & P.) Pers. (Leaves) Calea uniflora (N. R.) N. R. In vitro (Pro.) L. major Schmidt et al., 2012a Chersodoma jodopappa (Sch. Bip.) Ethanol N. R. L. amazonensis Rocha et al., 2005 Cabrera (Leaves + Stem) L. braziliensis L. donovani Chromolaena hirsute (N. R.) N. R. In vitro (Pro.) L. amazonensis Schmidt et al., 2012a lorentzii Griseb Ethanol N. R. L. amazonensis. Rocha et al., 2005 (Leaves + Stem) L. braziliensis L. donovani Echinacea purpurea Moench (Entire Saponin N. R. Leishmania sp. Rocha et al., 2005 plant) Eupatorium perfoliatum (N. R.) N. R. In vitro (axenic Ams.) L. donovani Schmidt et al., 2012a

Inula montana L. (Aerial parts) Methanol N. R. L. infantum Rocha et al., 2005 Jasonia glutinosa DC. (Aerial parts) Acetone In vitro (Pro.) L. donovani Rocha et al., 2005 Schmidt et al., 2012a Lychnophora markgravii (N. R.) N. R. In vitro (Ams.) L. amazonensis Schmidt et al., 2012a Munnozia fournetii H. Robinson Ethanol N. R. L. amazonensis Rocha et al., 2005 (Leaves + Stem) L. braziliensis L. donovani Neurolaena lobata Ethanol N. R. L. mexicana Berger et al., 2001 R. Br. (Leaves) L. braziliensis Ophryosporus piquerioides (DC.) Benth Ethanol N. R. L. amazonensis Rocha et al., 2005 (Entire plant) L. braziliensis Perezia multiflora Less. (H. & B.) Less L. donovani (Leaves) Pterocaulon alopecuroideum (Lam.) DC. (Entire plant) Senecio clivicolus Wedd.(Leaves +Stem) Stevia yaconensis Hieron. (Entire plant)

17

Table 1. Plants reported as antileishmanial (Continued…) Botanical source Extract/Fraction Bioactivity/life stage Leishmania spp References Asteraceae Chloroform In vitro (Ams.) L. aethiopica Carvalho and Ferreira, Vernonia amygdalina Delile (N. R.) 2001 Vernonia brachycalyx (Roots) N. R. In vitro (Pro.) L. major Schmidt et al., 2012a Carvalho and Ferreira, 2001 Vernonia squamulosa Hook. & Arn. Ethanol N. R. L. amazonensis Rocha et al., 2005 (Stem) L. braziliensis Werneria nubigena H.B. K. L. donovani (Leaves + Stem) Xanthium catharticum L. (Roots + Stem) Berberidaceae Alkaloid frction N. R. L. amazonensis Rocha et al., 2005 Berberis boliviana Lechl. (Bark + Stem) L. braziliensis Berberis bumeliaefolia Schum (Bark) L. donovani Berberis cf. laurina Epl. (Stem) Betulaceae N. R. In vitro (Pro.) L. major Schmidt et al., 2012b Betula species (N. R.) Berberis aff. Paucidentata Rusby Alkaloid frction. N. R. L. amazonensis Rocha et al., 2005 (Stem bark) L. braziliensis Bignoniaceae N. R. In vitro & in vivo L. amazonensis Rocha et al., 2005 Jacaranda copaia D. Don (Leaves) (Ams.) Tabebuia avellanedae (N. R.) N. R. In vitro (Pro.) L. braziliensis Schmidt et al., 2012a Kigelia pinnata DC. (Root bark) N. R. N. R. L. major Rocha et al., 2005 Bombacaceae Methanol N. R. L. panamensis Rocha et al., 2005 Huberodendron patinoi Cuatrec (Bark) Burseraceae N. R. N. R. L. donovani Schmidt et al., 2012b Boswellia serrata (N. R.) Protium amplum Cuatrec (Fruits) DCM N. R. L. amazonensis Rocha et al., 2005 L. braziliensis L. infantum Key: c: Data incomplete derived from an abstract; Pro.: promastigote; Ams.: amastigotes; DCM: dicloromethane; N. R.: not reported

18

Table 1. Plants reported as antileishmanial (Continued…) Botanical source Extract/Fraction Bioactivity/life stage Leishmania spp References Buxaceae N. R. In vitro (N. R.) L. major Schmidt et al., 2012a Sarcococca hookeriana (Whole air-dried plant) Calophyllaceae N. R. In vitro (Pro.& Ams.) L. amazonensis Schmidt et al., 2012a Calophyllum brasiliense (N. R.) Kielmeyera albopunctata (N. R.) N. R. In vitro (Pro.) L. donovani Schmidt et al., 2012a Cannabaceae N. R. In vitro (Pro.). L. donovani Schmidt et al., 2012a Cannabis sativa (N. R.) Caparraceae N. R. N. R. L. major Rocha et al., 2005 Capparis spinosa L. (Branches) Chloranthaceae N. R. In vitro (axenic Ams.) L. amazonensis. Schmidt et al., 2012b Hedyosmum angustifolium (N. R.) L. infantum Celastraceae N. R. N. R. L. donovani Schmidt et al., 2012b Austroplenckia populnea (N. R.) Maytenus senegalensis (Lam.) Exell DCM In vitro (Pro.). L. major El Tahir et al., 1998 (Stem bark) Salacia madagascariensis (N. R.) N. R. N. R. L. donovani Schmidt et al., 2012b Cistaceae N. R. In vitro (Pro.) L. donovani Schmidt et al., 2012b Cistus creticus (N. R.) Clusiaceae N. R. In vitro (Ams.) L. infantum Schmidt et al., 2012a Garcinia Livingstonei (N. R.) Garcinia lucida (Stem bark) DCM/ methanol In vitro (axenic Ams.) L. donovani Schmidt et al., 2012a (1 :1) Marila laxiflora Rusby (Leaves) DCM N. R. L. amazonensis Rocha et al., 2005 L. braziliensis Combretaceae N. R. In vitro (Pro.) L. amazonensis Schmidt et al., 2012b Combretum leprosum (N. R.) Crassulaceae N. R. In vitro (Pro.) L. tropica Schmidt et al., 2012b Aeonium lindleyi (N. R.) L. braziliensis Bryophyllum pinnatum Kurz (Leaves) Aqueus N. R. L. amazonensis Rocha et al., 2005 Kallanchoe pinnata (Kp) Bryophyllum Aqueus In vitro (Pro.) L. amazonensis Da Silva et al., 1995 pinnatum Kurz (synoname) (Leaves) Key: c: Data incomplete derived from an abstract; Pro.: promastigote; Ams.: amastigotes; DCM: dicloromethane; N. R.: not reported

19

Table 1. Plants reported as antileishmanial (Continued…) Botanical source Extract/Fraction Bioactivity/life stage Leishmania spp References Cupressaceae N. R.. In vitro (Pro.) L. donovani Schmidt et al., 2012b Juniperus procera (N. R.) Dilleniaceae Chloroform N. R. L. amazonensis Rocha et al., 2005 Doliocarpus dentatus Kubitzki (Stem) Ebenaceae N. R. In vitro (Pro.) & L. donovani Schmidt et al., 2012a Diospyros Montana (Bark) in vivo (Ams.) Hazra et al., 1995

Euphorbiaceae Alcohol (N. R) N. R. L. donovani Carvalho and Ferreira, Anthostema senegalense A. Juss 2001 (N. R.) Celaenodendron mexicanum (N.R.) N. R. In vitro (Pro.) L. donovani Schmidt et al., 2012b Pera benensis Rusby Ethanol N. R. L. amazonensis Rocha et al., 2005 (Root bark + stem bark) L. braziliensis L. donovani Ricinus communis V. A. Moshkin N. R. N. R. L. major Rocha et al., 2005 (Branches) Illiciaceae N. R. In vitro (axenic Ams.) L. donovani Schmidt et al., 2012a Illicium floridanum (N. R.) Fabaceae N. R. In vitro (Pro.&Ams.) L. braziliensis Schmidt et al., 2012a Amburana cearensis (N. R.) L. donovani L. amazonensis Astragalus bicuspis (N. R.) N. Rp. In vitro (Pro.) L. major Schmidt et al., 2012b Caesalpinia echinata (N. R.) N. R. In vitro/ams. L. amazonensis Schmidt et al., 2012b Canavalia brasiliensis (Seeds) N. R. In vitro (Pro.) & L. amazonensis Barral-Netto et al., 1996 in vivo (Ams.) Crotalaria barbata R. Gach Ethanol N. R. L. donovani Rocha et al., 2005 (Entire plant) Desmodium gangeticum L. (Leaves) Methanol N. R. L. donovani Rocha et al., 2005 Glycyrrhiza sp. (N. R.) N. R. In vitro (Pro.) L. donovani Schmidt et al., 2012a Lonchocarpus sp. (N. R.) N. R. In vitro (Pro.) L. braziliensis Schmidt et al., 2012a L. amazonensis L. donovani

20

Table 1. Plants reported as antileishmanial (Continued…) Botanical source Extract/Fraction Bioactivity/life stage Leishmania spp References Fabaceae N. R. In vitro (Pro.) L. mexicana Schmidt et al., 2012a Lonchocarpus xuul (N. R.) Machaerium multiflorum (N. R.) N. R. In vitro (Pro.) L. donovani Schmidt et al., 2012a Periandra mediterranea Taub. (Roots) Saponin fraction N. R. L. donovani Rocha et al., 2005 Prosopis glandulosa var. (Leaves) N. R. In vitro (Pro.) L. donovani Schmidt et al., 2012a Psorothamnus polydenius (N. R.) N. R. In vitro (axenic Ams.) L. donovani Schmidt et al., 2012a Spartium junceum L. (Branches) N. R. N. R. L. major Rocha et al., 2005 Gentianaceae Ethanol In vivo (Ams.) L. donovani Rocha et al., 2005 Swertia chirata Buch. Ham.Ex Wall. Carvalho and Ferreira, (Entire plant) 2001 Geraniaceae Ethanol Invitro (Ams.) L. donovani Rocha et al., 2005 Pelargonium sidoides DC. (c) Schmidt et al., 2012a Lamiaceae N. R. In vitro (Pro.) L. major Schmidt et al., 2012a Perovskia abrotanoides (N. R.) Phlomis brunneogaleata (N. R.) N. R. N. R. L. donovani Schmidt et al., 2012a Ocimum sanctum (N. R.) N. R. In vitro (Pro.). L. major Schmidt et al., 2012a Lauraceae Ethyl acetate N. R. L. amazonensis Rocha et al., 2005 Aniba canelilla H. B. K. (Stem) L. braziliensis Aniba species (Stem) Ethanol N. R. L. amazonensis Rocha et al., 2005 L. braziliensis Ocotea duckei (N. R.) N. R. In vitro (Pro.) L. chagasi Schmidt et al., 2012a L. infantum Ocotea lancifolia (N. R.) Alkaloid crude In vitro (Pro.) L. amazonensis Schmidt et al., 2012a extract L. braziliensis L. donovani Liliaceae (c) In vivo (Ams.) L. major Rocha et al., 2005 Allium sativum L. (N. R.) Asparagus africanus Lam. (N. R.) N. R. In vitro (Pro.) L. major Carvalho and Ferreira, 2001 Key: c: Data incomplete derived from an abstract; Pro.: promastigote; Ams.: amastigotes; DCM: dicloromethane; N. R.: not reported

21

Table 1. Plants reported as antileishmanial (Continued…) Botanical source Extract/Fraction Bioactivity/life stage Leishmania spp References Liliaceae Methanol N. R. Leishmania sp. Carvalho and Ferreira, arborea Hort. Angl. 2001 (Seed pulp) Dracaena. mannii Baker(Seed pulp) Leguminosae DCM In vitro (N. R.) L. amazonensis Bravo B. et al., 1999 Amburana cearensis A.C Smith. L braziliensis (Stem bark) L.donovani

Centrolobium sclerophyllum (Wood) Chloroform In vitro (Pro.) L. amazonensis Araujo et al., 1998 Maliaceae DCM N. R. L. amazonensis Rocha et al., 2005 Guarea polymera Little (Leaves) L. braziliensis Malpighiaceae N. R. In vitro (Ams.) L. amazonensis Schmidt et al., 2012b Lophanthera lactescens (N. R.) Malvaceae N. R. N. R. L. major Rocha et al., 2005 Malva nicaeensi All. (Branches) Melastomaceae Ethanol N. R. L. donovani Rocha et al., 2005 Tibouchina semidecandra Cogn. (Aerial parts) Meliaceae Methanol In vitro (Pro.) L. major El Tahir et al., 1998 Azadirachta indica A. Juss. (Stem bark) Guarea rhophalocarpa (N. R.) N. R. In vitro (Pro.&Ams.) L. donovani Schmidt et al., 2012a Khaya senegalensis A. Juss (N. R.) Alcohol (N. R) N. R. L. donovani Rocha et al., 2005 Khaya senegalensis (N. R.) N. R. In vitro (Pro.) L. major Schmidt et al., 2012b Menispermaceae Alkaloid fraction N. R. L. amazonensis Rocha et al., 2005 Abuta pahni Mart. (Stem) L. braziliensis Abuta rufescens Aublet (Bark) Alkaloid fraction N. R. L. amazonensis Rocha et al., 2005 Anomospermum bolivianum K ruk. & Alkaloid fraction N. R. L. amazonensis Rocha et al., 2005 Mold (Bark) L. braziliensis Stephania dinklagei (Aerial parts) N. R. In vitro (Pro.&Ams.) L. donovani Chan-Bacab and Peña- Rodríguez, 2001 Key: c: Data incomplete derived from an abstract; Pro.: promastigote; Ams.: amastigotes; DCM: dicloromethane; N. R.: not reported

22

Table 1. Plants reported as antileishmanial (Continued…) Botanical source Extract/Fraction Bioactivity/life stage Leishmania spp References Menispermaceae Ethanol N. R. L. donovani Rocha et al., 2005 Tinospora cordifolia (Wild) Hook.f. & Thoms (Stem) Moraceae Aqueus N. R. L. donovani Rocha et al., 2005 Dorstenia multiradiata Engl. (Leaves) Myristicaceae DCM N. R. L. amazonensis Rocha et al., 2005 Otoba novogranatensis Moldenke L.braziliensis (Leaves) L.infantum Otoba novogranatensis Moldenke Methanol N. R. L. amazonensis Rocha et al., 2005 (Leaves) L. braziliensis L. infantum Otoba parvifolia (mgf.) A. H. Gentry DCM N. R. L. amazonensis Rocha et al., 2005 (Bark) L. braziliensis Virola pavonis (Leaves) N. R. In vitro (Pro.&Ams.) L. donovani Barata et al., 2000 Virola surinamensis (Leaves) Myrsinaceae Ethanol N. R. L. braziliensis Rocha et al., 2005 Myrsine pellucida Spreng (Stem bark) Nepenthaceae N. R. In vitro (Pro.) & L. amazonensis Schmidt et al., 2012a Nepenthes thorelii (N. R.) in vivo (Ams.) L. donovani L. braziliensis L. mexicana. L.venezuelensis Olacaceae N. R. In vitro (N. R.) L. major Schmidt et al., 2012a Minquartia guianensis (Stem bark) Oleaceae N. R. In vitro & in vivo (N. L. donovani Rocha et al., 2005 Nyctanthes arbortristis L. (N. R.) R.) Carvalho and Ferreira 2001 Orobanchaceae N. R. N. R. L. donovani Schmidt et al., 2012a Melampyrum arvense (N. R.) Oxalidaceae N. R. In vitro (Pro.) L. amazonensis Schmidt et al., 2012a Oxalis erythrorhiza (N. R.) L. donovani Papaveraceae Alkaloid fraction. N. R. L. amazonensis Rocha et al., 2005 Bocconia integrifolia H & B L. braziliensis (Leaves + Stem bark) L. donovani

23

Table 1. Plants reported as antileishmanial (Continued…) Botanical source Extract/Fraction Bioactivity/life stage Leishmania spp References Papaveraceae Alkaloid fraction N. R. L. amazonensis Rocha et al., 2005 Bocconia pearcei Hutch. (Leaves) L. braziliensis L. donovani Phrymaceae N. R. In vitro (axenic Ams.) L. donovani Schmidt et al., 2012a Mimulus bigelovii (N. R.) Phytolaccaceae Butanol N. R. L. enriettii Rocha et al., 2005 Phytolacca dodecandra L’Herit (Fruits) Piperaceae Ethanol N. R. L. amazonensis Rocha et al., 2005 Peperomia galioides H. B. & K. (Entire plant) Peperomia galioides H. B. & K Ethanol N. R. L. braziliensis Rocha et al., 2005 (Entire plant) Petroleum ether L. chagasi L. donovani Peperomia galioides H. B. & K. (N. R.) N. R. In vitro (Pro.) L. braziliensis Chan-Bacab and Peña- L. amazonensis Rodríguez, 2001 L. donovani Piper aduncum L. Petroleum ether In vitro (Pro.&Ams.) L. amazonensis Rocha et al., 2005 Schmidt et al., 2012a Piper elongatum (N. R.) N. R. In vitro (Pro.) L. braziliensis Schmidt et al., 2012a L. infantum L. tropica Piper nigrum (Fruits) N. R. In vitro (Pro.) L. donovani Schmidt et al., 2012a L. amazonensis Piper rusbyi C. DC. (Entire plant) Ethyl acetate In vitro (Pro.) L. amazonensis Rocha et al., 2005 L. braziliensis L. donovani Schmidt et al., 2012a

In vivo (Ams.) L. amazonensis Piper sanguineispicum (Leaves) N. R. In vitro (axenic Ams.) L. amazonensis Schmidt et al., 2012a Plagiochilaceae N. R. In vitro (Ams.) L. amazonensis Schmidt et al., 2012b Plagiochila disticha (N. R.) Pleosporaceae N. R. In vitro (Ams.) L. amazonensis Schmidt et al., 2012a Cochliobolus sp. (N. R.)

24

Table 1. Plants reported as antileishmanial (Continued…) Botanical source Extract/Fraction Bioactivity/life stage Leishmania spp References Ranunculaceae N. R. In vitro (Pro.) L. braziliensis Schmidt et al., 2012a Consolida oliveriana (N. R.) L. peruviana Thalictrum flavum (Roots) N. R. N. R. L. major Schmidt et al., 2012a Rubiaceae N. R. N. R. L. donovani Schmidt et al., 2012a Carapichea ipecacuanha (Roots) Faramea guianensis (Aublet.) Bremek Aqueus In vitro & in vivo L. amazonensis Rocha et al., 2005 (Leaves) (Ams.) Carvalho and Ferreira., 2001 Hintonia latiflora (N. R.) N. R. In vitro (Pro.) L. donovani Schmidt et al., 2012a Nauclea latifolia (Root bark) Chloroform In vitro (axenic Ams.) L. donovani Schmidt et al., 2012a Rutaceae Ethyl acetate N. R. L. amazonensis Rocha et al., 2005 Dictyoloma peruvianum Planch Alkaloid fraction L. braziliensis (Stem bark) Haplophyllum bucharicum (N. R.) N. R. In vitro (Pro.&Ams.) L. infantum Schmidt et al., 2012a Helietta apiculata (Bark) N. R. In vitro (Pro.) & L. amazonensis Schmidt et al., 2012a in vivo (Ams.) L. braziliensis L. infantum Galipea longiflora Kr. Alkaloid fraction In vitro (Pro.) & L. amazonensis Carvalho and Ferreira., (Root bark + Stem bark + Leaves) in vivo (Ams.) L. braziliensis 2001 L. donovani L. venezuelensis Raputia heptaphylla Pittier (Leaves) N. R. In vitro (Pro.) L. panamensis Schmidt et al., 2012a Swinglea glutinosa Merr. (Bark) DCM N. R. L. amazonensis Rocha et al., 2005 L. braziliensis L. infantum Thamnosma rhodesica (N. R.) N. R. In vitro (Pro.) L. major Schmidt et al., 2012a Sapindaceae Ethanol N. R. L. amazonensis Rocha et al., 2005 Nicotiana glauca Grahm. L. braziliensis (Leaves + stem) L. donovani Key: c: Data incomplete derived from an abstract; Pro.: promastigote; Ams.: amastigotes; DCM: dicloromethane; N. R.: not reported

25

Table 1. Plants reported as antileishmanial (Continued…) Botanical source Extract/Fraction Bioactivity/life stage Leishmania spp References Sapindaceae Ethanol N. R. L. amazonensis Rocha et al., 2005 Serjania tenuifolia Radlk (Leaves +stem) L. braziliensis Solanum actaeabotrys Rusby (Leaves) Ethanol N. R. L. braziliensis Rocha et al., 2005 L. donovani Scrophulariaceae DCM N. R. L. amazonensis Rocha et al., 2005 Conobea scoparioides (Cham.&Schltd.) L. braziliensis Benth (Leaves) Picrorhiza kurroa Royle, ex Benth Ethanol In vivo (Ams.) L. donovani Carvalho and Ferreira, (Roots + rhizome) 2001 Scrophularia scorodonia L. (Leaves) Methanol N. R. L. infantum Rocha et al., 2005 Solanaceae Ethanol In vitro (Pro.) L. amazonensis Rocha et al., 2005 Saracha punctata Ruiz & Pav. (Leaves) L. braziliensis Carvalho and Ferreira, L. donovani 2001 Solanum actaeabotrys Rusby (Leaves) Ethanol N. R. L. amazonensis Rocha et al., 2005 Solanum luteum Mill (Branches) N. R. N. R. L. major Rocha et al., 2005 Sterculiaceae Chloroform N. R. L. donovani Rocha et al., 2005 Cola attiensis Aubrev. & Pellegr. (Seeds) Ulmaceae Chloroform In vitro (Pro.) & L. amazonensis Rocha et al., 2005 Ampelocera edentula Kuhlm (Stem bark) in vivo (Ams.) L. venezuelensis Carvalho and Ferreira 2001 Verbenaceae Ethanol N. R. L. donovani Rocha et al., 2005 Nyctanthes arbortristis L. (Aerial parts) Vitex heterophylla Miq. (Leaves) Ethanol N. R. L. donovani Rocha et al., 2005 Winteraceae N. R. In vitro (Pro.) L. amazonensis Schmidt et al., 2012b Drimys species (N. R.) L. braziliensis L. chagasi Zingiberaceae N. R. In vitro (Pro.) L. donovani Schmidt et al., 2012b Aframonum sceptrum (N. R.) Zygophyllaceae (Nitrariaceae) N. R. In vitro (N. R.) L. donovani Schmidt et al., 2012a Larrea tridentata (N. R.) Peganum harmala (Seeds) N. R. In vitro (Pro.) & L. donovani Schmidt et al., 2012a in vivo (Ams.) Key: c: Data incomplete derived from an abstract; Pro.: promastigote; Ams.: amastigotes; DCM: dicloromethane; N. R.: not reported

26

2.2.3 Alkaloids from medicinal plants reported to exhibited antileishmanial activity Alkaloids reported to have antileishmanial activity from medicinal plants including diverse chemical structures, such as: quinolines, isoquinolines, indoles, diterpenoids, steroidal and amide alkaloids. Table 2 compiles the names, the antileishmanial activity of these alkaloids against various leishmania species, and their toxicity. The exact chemical structures of these alkaloids are presented in Figure 2.

27

Table 2. Reported antileishmanial alkaloids

QUINOLINE ALKALOIDS Compounds/botanical source Bioactivity/life Leishmania sp. Toxicity References stage γ-Fagarine 1 In vitro (Pro.) & L.amazonensis N. R. Schmidt et al., 2012b Helietta apiculata (Rutaceae) in vivo (Ams.) L. braziliensis L. infantum N-methyl-8-methoxyflindersine 2 In vitro (Pro.) L. panamensis N. R. Schmidt et al, 2012b Raputia heptaphylla (Rutaceae) Chimanine A 3 In vitro (Pro.) & L. braziliensis N. R. Carvalho and Ferreira, in vivo (Ams.) L. amazonensis 2001 Chimanine B 4 L. venezuelensis In vitro (Pro.) L.braziliensis Chimanine D 5 L. amazonensis Calipea longiflora (Rutaceae) In vivo (Ams.) L. venezuelensis Rocha et al., 2005 L. donovani Key: Pro.: promastigote; Ams.: amastigotes; N. R.: not reported

28

Table 2. Reported antileishmanial alkaloids (continued…)

ISOQUINOLINE ALKALOIDS Compounds/botanical source Bioactivity/life Leishmania sp. Toxicity References stage (-)-Coreximine 6 N.R. L. amazonensis N.R. Rocha et al., 2005 Isoguattouregidine 7 L. donovani Carvalho and Ferreira, Guatteria foliosa 2001 (Annonaceae) Caaverine 8 In vitro (Pro.) L. amazonensis Caaverine, Schmidt et al., 2012b Nordomesticine 9 L.braziliensis hepatotoxic Ocotea lancifolia (Lauraceae) L. donovani Berberine 10 In vitro (Pro.) & L. donovani Rocha et al., 2005 Palmatine 11 in vivo (Ams.) L. major Chan-Bacab and Peña- Jasonia glutinosa (Asteraceae) L. panamensis Rodríguez, 2001 and other several Leishmania sp. Duguetine 12 In vitro (Pro.) L. braziliensis Duguetine and Schmidt et al., 2012b β-N-oxide duguetine 13 its β -N- Dicentrinone 14 oxideshowed N-methylglaucine 15 considerable Duguetia furfuracea cytotoxicity against three (Annonaceae) cancer cell lines. For (14) and (15) N.R Key: Pro.: promastigote; Ams.: amastigotes; N. R.: not reported

29

Table 2. Reported antileishmanial alkaloids (continued…)

ISOQUINOLINE ALKALOIDS Compounds/botanical source Bioactivity/life Leishmania sp. Toxicity References stage Xylopine 16 N. R. L. panamensis Showed Schmidt et al., 2012b Guatteria amplifolia L. mexicana higher (Annonaceae) toxicity towards L. mexicana than to macrophages (host cells of leishmania spp.) Emetine 17 N. R. L. donovani High acute Schmidt et al., 2012b (-) Emetine & 2,3-dehydro and sub acute emetine L. tropica toxicity Rocha et al., 2005 Carapichea ipecacuanha (Rubiaceae) Key: Pro.: promastigote; Ams.: amastigotes; N. R.: not reported

30

Table 2. Reported antileishmanial alkaloids (continued…)

NAPHTHYLISOQUINOLINE ALKALOIDS Compounds/botanical source Bioactivity/life Leishmania sp. Toxicity References stage Dioncophylline C 18 In vitro (Pro.) L. major 18, 19, 20 and Schmidt et al, 2012b Ancistroheynine B 19 21inhibited the Entdioncophylleine A 20 growth of N-phenyl-6,8-dimethoxy-1,3- macrophage dimethylisoquinolinium cell line J774.1. at chloride 21 concentrations Ancistrocladiniums A 22 similar to Ancistrocladus those needed sp.(Ancistrocladaceae) to inhibit the Ancistrocladiniums B 23 proliferation of Synthetic analogue 24 (name parasite. N.R.) 23 and 24 were toxic against the macrophage cell line at concentrations significantly higher than those needed to inhibit parasite cell growth. Key: Pro.: promastigote; Ams.: amastigotes; N. R.: not reported

31

Table 2. Reported antileishmanial alkaloids (continued…)

NAPHTHYLISOQUINOLINE ALKALOIDS Compounds/botanical source Bioactivity/life Leishmania sp. Toxicity References stage 6,4'-O- In vitro (Ams.) L. donovani N.R Schmidt et al., 2012b demethylancistrocladinium A & L. major 25 in vitro (Pro.) Ancistrocladus cochinchinensis (Ancistrocladaceae) 26, 27, 28 and 29 N. R. L. donovani N.R. Schmidt et al., 2012b Ancistrocladus Taxon (Ancistrocladaceae) Key: Pro.: promastigote; Ams.: amastigotes; N. R.: not reported

32

Table 2. Reported antileishmanial alkaloids (continued…)

BISBENZYLISOQUINOLINE ALKALOIDS Compounds/botanical source Bioactivity/life Leishmania sp. Toxicity References stage Thalfoetidine 30 N.R. L. major N.R. Schmidt et al, 2012b Northalfoetidine 31 No high Thalictrum flavum cytotoxicity (Ranunculaceae) (cell typeN.R.) Isotetrandrine 32 In vivo (Ams.) L.amazonensis N.R. Fournet et al., 1993 Limaciopsis loangensis L. venezuelensis (Menispermaceae) Antioquine 33 Pseudoxandra scferocarpa(Annonaceae) Berbamine 34 Berberis bofiuiana (Berberidaceae) Gyrocarpine 35 Gyrocarpus arnericanus (Hernandiaceae) Key: Pro.: promastigote; Ams.: amastigotes; N. R.: not reported

33

Table 2. Reported antileishmanial alkaloids (continued…)

BENZO[C]PHENANTHRIDINE ALKALOIDS Compounds/botanical source Bioactivity/life Leishmania sp. Toxicity References stage Dihydro-chelerythrine 36 In vitro (axenic L. donovani No toxicity Schmidt et al., 2012b 6-acetonyldihydrochelerythrine37 Ams.) on Vero cells Lucidamine A 38 Lucidamine B 39 Garcinia lucida (Clusiaceae)

QUINAZOLINE ALKALOIDS Peganine hydrochloride In vitro (Pro.) & L. donovani N.R. Schmidt et al, 2012b dihydrate 40 in vivo (Ams.) Peganum harmala (Nitrariaceae) formerly (Zygophyllaceae) Key: Pro.: promastigote; Ams.: amastigotes; N. R.: not reported

34

Table 2. Reported antileishmanial alkaloids (continued…)

INDOLE ALKALOIDS Compounds/botanical Bioactivity/life Leishmania sp. Toxicity References source stage Harmaline 41 N. R. Leishmania sp. N.R. Chan-Bacab and Peña- Peganum harmala Rodríguez, 2001 (Nitrariaceae) & Passiflora Incarnate (Passifloraceae) Coronaridine 42 In vitro (Pro.) & L.amazonensis cytotoxicity on Delorenzi et al., 2001 macrophages Peschiera australis in vivo (Ams.) mammalian cells, (Apocynaceae) murine (20 and 10 μg/mL for 24 h.) and human (20 μg/mL for 7 days) showed good viability of cells, but higher doses on human (40 μg/mL) showed toxcicity after 7 days. Ellipticine 43 In vitro (axenic L. donovani Very high Schmidt et al., unpublished Aspidosperma vargasii Ams.) cytotoxicity (Apocynaceae) against L6 cells. Strictosamide 44 In vitro (axenic L. donovani Cytotoxicity Khalid, 2012 Nauclea latifolia (Rubiaceae) Ams.) 51.9 μg/mL indicates some selectivity Key: Pro.: promastigote; Ams.: amastigotes; N. R.: not reported

35

Table 2. Reported antileishmanial alkaloids (continued…)

BISINDOLE ALKALOIDS Compounds/botanical source Bioactivity/lif Leishmania sp. Toxicity References e stage Conoduramine 45 In vivo (Ams.) L.amazonensis Weak Carvalho and Ferreira, Conodurine 46 toxicity 2001 Gabunine (N-demethyl towards Rocha et al., 2005 Conodurine) 47 macrophage Peschiera uan heurkii Muell. host cells Arg.L. Allorge Tabernaemontana uan heurkii Muell. Arg. (Syn.) (Apocynaceae)

INDOLIZIDINE ALKALOIDS Prosopilosidine 48 In vitro (Pro.) L. donovani Cytotoxicity against Schmidt et al., 2012b selected human Prosopis glandulosa var. cancer cell lines, (Fabaceae) SK-MEL, KB, BT 549 and SK-OV-3, Prosopilosine 49 48 and 50were Isoprosopilosine 50, 51 and 52 weakly active towards all of these (Source N.R.) cancer cell lines but un toxic to Vero, monkey kidney fibroblast cells and LLCPK, pig kidney epithelia) cells. Key: Pro.: promastigote; Ams.: amastigotes; N. R.: not reported

36

Table 2. Reported antileishmanial alkaloids (continued…)

DITERPENOID ALKALOIDS Compounds/botanical source Bioactivity/lif Leishmania sp. Toxicity References e stage Azitine 53 In vitro (Pro.) L. infantum N. R. Schmidt et al., 2012b Isoazitine 54 15,22-O-diacetyl-19- oxodihydroatisine 55 Ranunculaceae (Plant N.R.) Key: Pro.: promastigote; Ams.: amastigotes; N. R.: not reported

37

Table 2. Reported antileishmanial alkaloids (continued…)

STEROIDAL ALKALOIDS Compounds/botanical source Bioactivity/life Leishmania sp. Toxicity References stage Hookerianamide J 56 In vitro (stage L. major N.R. Schmidt et al., 2012b Hookerianamide K 57 N.R.) 58, 59, 60, 61, 62, 63, 64 and 65 Sarcococca hookeriana (Buxaceae) Holacurtine 66 N.R. L. donovani N.R. Rocha et al., 2005 N-demethylholacurtine 67 17-epiholacurtine 68 17-epi-N-demethyl holacurtine Halocurtinol 69 Holamine 70 Holarrhena curtisii (Apocynaceae) Sarachine 71 Saracha punctata In vitro (Pro.) L. chagasi N.R. Carvalho and Ferreira, 2001 (Solanaceae)Annona L. braziliensis senegalensis(Annonaceae) N.R. L. major Rocha et al., 2005 Key: Pro.: promastigote; Ams.: amastigotes; N. R.: not reported

38

Table 2. Reported antileishmanial alkaloids (continued…)

AMIDE ALKALOIDS Compounds/botanical source Bioactivity/life Leishmania sp. Toxicity References stage Piperine 72 In vitro (Pro.) L. donovani N. R. Schmidt et al., 2012b Piper nigrum (Piperaceae) L. amazonensis 2-Benzoxazolinone 73 In vitro (Pro.) L. donovani N. R. Carvalho and Ferreira, 2001 Acanthus illicifolius L. (Acanthaceae) Dictyolamide A 74 In vitro (Pro.) L.amazonensis N. R. Carvalho and Ferreira, 2001 Dictyolamide B 75 L. braziliensis Dictyoloma peruvianum Planch. (Rutaceae) (Simarubaceae) Liriodenine 76 & Unonopsine 77 In vitro (Pro.) L. donovani Liriodenine Rocha et al., 2005 Unonopsis buchtienii R. E. Fries L. major showed high Carvalho and Ferreira, 2001 (Annonaceae) cytotoxicity, Liriodenine 76 L. amazonensis against Vero Rollinia emarginata L. braziliensis cell lines. Schdl.(Anonnaceae) L. donovani Key: Pro.: promastigote; Ams.: amastigotes; N. R.: not reported

39

γ-Fagarine 1 N-methyl-8-methoxyflindersine 2 Chimanine A 3 Chimanine B 4

Chimanine D 5 (-)-Coreximine 6 Isoguattouregidine 7 Caaverine 8

Nordomesticine 9 Berberine 10 Palmatine 11 Duguetine 12

Figure 2. Chemical structures of reported antileishmanial alkaloids

40

β-N-oxide duguetine 13 Dicentrinone 14 N-methylglaucine 15 Xylopine 16

Emetine 17 Dioncophylline C 18 Ancistroheynine B 19 Entdioncophylleine A 20

N-phenyl-6,8-dimethoxy- 1,3-dimethylisoquinolinium chloride 21 Ancistrocladiniums A 22 Ancistrocladiniums B 23 Synthetic analogue 24 (from 22 name N.R) Figure 2. Chemical structures of reported antileishmanial alkaloids (continued …)

41

6,4'-O-demethylancistrocladinium A 25 26 27 28

29 Thalfoetidine 30 Northalfoetidine 31

Isotetrandrine 32 Antioquine 33 Berbamine 34

Figure 2. Chemical structures of reported antileishmanial alkaloids (continued …)

42

Gyrocarpine 35 Dihydro-chelerythrine 36 6-acetonyldihydrochelerythrine 37

Lucidamine A 38 Lucidamine B 39 Peganine hydrochloride dehydrate 40 Harmaline 41

Coronaridine 42 Ellipticine 43 Strictosamide 44

Figure 2. Chemical structures of reported antileishmanial alkaloids (continued …)

43

Conoduramine 45 Conodurine 46 Gabunine 47 (N-demethylConodurine)

Prosopilosidine 48 Prosopilosine 49 Isoprosopilosine 50 51

Figure 2. Chemical structures of reported antileishmanial alkaloids (continued …)

44

52 Azitine 53 Isoazitine 54 15,22-O-diacetyl-19-oxodihydroatisine 55

Hookerianamide J 56 Hookerianamide K 57 ∆16,17 58 ∆4,5 & 14,15

59 ∆2,3 60 61

Figure 2. Chemical structures of reported antileishmanial alkaloids (continued …)

45

62 63 ∆5,6 64

65 ∆2,3 & Holacurtine 66 N-demethylholacurtine 67

Figure 2. Chemical structures of reported antileishmanial alkaloids (continued …)

46

17-epiholacurtine 68 Halocurtinol 69 Holamine 70

Sarachine 71 Piperine 72 2-Benzoxazolinone 73 Dictyolamide A 74

Dictyolamide B 75 Liriodenine 76 Unonopsine 77

Figure 2. Chemical structures of reported antileishmanial alkaloids (continued …)

47

2.3 Plants investigated in the present study Two plants were investigated in this study, Argemone mexicana L. and Nauclea latifolia Smith. The number of reported isolated compounds from A. mexicana and N. latifolia are 95 and 248 compunds, respectively (DNP database, 2011).

2.3.1 Argemone mexicana L. (Papaveraceae) 2.3.1.1 The botanical description of Argemone mexicana L. A. mexicana L. has different vernacular names, such as mexican prickly poppy, mexican poppy, devil’s fig, golden thistle of Peru. The species originates from Mexico but is now a worldwide distributed tropical weed, especially in coastal regions, and widely naturalized in the Old World (Scott, 1996). Argemone mexicana counts fourteen synonyms: Echtrus trivialis Lour., Echtrus mexicanus (L.) Nieuwl., Argemone vulgaris Spach, A. versicolor Salisb., A spinosa Moench, A. sexvalis Stokes, A. ochroleuca Sweet, A. mucronata Dum. Cours. ex Steud., A. mexicana var. typical Prain, A. mexicana var. parviflora Kuntze, A. mexicana var. ochroleuca (Sweet) Lindl., A. mexicana var. lutea Kuntze, A. mexicana fo. leiocarpa (Greene) G.B. Ownbey, A. leiocarpa Greene (MBG, 2009).

This annual plant can usually reach 60 – 90 cm in height. Stems are often branching at 2.5 – 8 dm from the base and branches are sparingly prickly, presenting bright yellow latex. Leaves are sessile, spiny and sinuate-pinnate. Flowers are large (4 – 7 cm of diameter), subtended by 1 – 2 foliaceous bracts; petals are bright yellow. Stamens are in number of 30 – 50. Fruits are

48 presented as prickly oblongovoid capsules, opening by 4 – 6 valves, containing numerous spherical seeds of 1.6 – 2 mm (Eichler, 1865).

Leaves are alternate, without stipules. Stems, leaves and other parts of the plant contain a well-developed system of secretory canals which produce yellow, milky or watery latex. The flowers are large and conspicuous, either solitary or arranged in cymose or racemose inflorescences (Heywood, 1993). The genus Argemone L. is closely allied to the genus Papaver L. and is mainly distributed in America (10 species). Species have white or yellow flowers similar to those of the genus Papaver, although the leaves of some Argemone species are prickly (Heywood, 1993).

The family Papaveraceae belongs to the order Papaverales within the Angiosperms classification. The family comprises ca. 250 species distributed within ca. 26 genera, most of them native to the northern temperate zone. Species are mainly herbaceous annuals or perennials but with some shrubs, all of which produce latex (Heywood, 1993). Economically the most important species in this family is Papaver somniferum (opium poppy) which yields opium. Seeds are used in baking and yield an important drying oil. Likewise, the seeds of Glaucium flavum and Argemone mexicana yield oils which are important in the manufacturing of soaps. Many species are cultivated as garden ornamental plants (Heywood, 1993).

49

2.3.1.2 Ethnopharmacological importance of Argemone mexicana L. The plant used as traditional medicine from ancient time. The word Argemone is derived from the Greek, argema meaning eye cataract, as the juice of this plant was used as a remedy in the treatment of this disease (Hakim, 1954; Diaz-Chavez, 2009). Different parts of the plant have been used in the traditional medicine for the treatment of fever, pain, diarrhea, cutaneous infections, itches, conjunctivitis and cancer. The oil from the seed is purgative, but prolonged ingestion produces toxic effect and even death (Diaz-Chavez, 2009).

Argemone mexicana has been used for more than 100 years in Indian traditional medicine to treat several diseases. A paste prepared from the roots is used for treating scorpion bite, the latex is used for eye inflammation and the juice of leaves as well as the latex with lemon juice is used for treating malaria (Kosalge and Fursule, 2009; Poonam and Singh, 2009).

In Mexico, the seeds are recommended as an antidote to snake venom. In India, the smokes of the seeds are used to relieve toothache. The protein– dissolving substances in the fresh yellow milky seeds extract are effective in the treatment of cold sore, cutaneous infections, warts, skin diseases, itches, dropsy and jaundice (Bhattacharjee et al., 2010).

Many reports were published showing various biological activities of different parts of the plant as well as pure constituents isolated from these parts. With concern to antiparasitic activity of A. mexicana, it is used as antimalarial in many African countries, including Sudan, Mali and .

50

Its in vitro efficacy against Plasmodium falciparum has been confirmed (Graz et al., 2009). Another study indicated the toxicity of seeds and leaves of the plant against three vector mosquitoes Culex quinquefasciatus, Anopheles stephensi and Aedes aegypti (Sakthivadivel and Daniel, 2008).

A mixture of quaternary alkaloids from Argemone mexicana showed in vitro activity against some phytopathogenic fungi (Singh et al., 2010a). Another study suggested that the roots could be a potential source of natural antioxidant that could have greater importance as therapeutic agent in preventing oxidative stress related degenerative diseases (Perumal et al., 2010).

Different studies showed that A. mexicana has in vitro antibacterial activity against some human pathogenic bacteria (Mohana et al., 2008; Singh et al., 2009; Reddy, 2010; Bhattacharjee et al., 2010). A study on leaves of A. mexicana justified its activity for wound healing. Aqueous extract of the leaves have been reported to exhibit anti-inflammatory activity. The alkaloid fraction of the roots is reported to possess anti-inflammatory activity and strong uterine stimulant effect (Dash and Murthy, 2011). In vitro study on the chloroform fraction of the whole plant showed growth inhibition against human nasopharyngeal carcinoma and human gastric cancer as well as anti- HIV activity (Chang et al., 2003b).

Previous chemical investigations of various parts of this plant revealed the occurrence of phenolic compounds and several alkaloids including, benzophinanthridine, protopine, benzylisoquinoline and berberine type. The isoquinoline type has been reported to possess anticholinergic and antihistaminic properties (Singh et al., 2010b; Piacente et al., 1997). The aerial

51 parts of Argemone mexicana, in the form of an infusion, are widely used in folk medicine for their analgesic properties (Piacente et al., 1997).

An ethnobotanical and retrospective treatment-outcome clinical study conducted in the Sikasso and Bandiagara regions of Mali showed that A. mexicana was one of the most effective plants used traditionally to treat non- complicated and severe malaria in that country (Diallo et al., 2006).

An entire research project has been developed by the non-governmental organization Antenna Technologies (Geneva) and the Department of Traditional Medicine (DTM) of Mali to establish the safety and efficacy of the traditional preparation of A. mexicana in order to develop an improved traditional medicine (ITM) or “medicament traditionnel amélioré” (MTA). In Mali, the DTM, as a part of the Ministry of Health, is responsible for the production of ITMs to be included in the National Formulary. As a preliminary result of this study, the decoction of A. mexicana was demonstrated to be active against malaria when taken twice a day. Higher doses (four times a day) were associated with a risk of cardiac toxicity.This suggested the presence of other active compounds. The search for other active compounds in the decoction was later considered (Willcox et al., 2007).

52

2.3.1.3 Phytochemistry of Argemone mexicana L. From the chemical point of view, several isoquinoline alkaloids have been isolated from A. mexicana extracts (Bentley, 1998; Chang et al., 2003a; Chang et al., 2003b; Israilov and Yunusov, 1986). The isoquinoline alkaloids are a large family of alkaloids derived mostly from the amino acid tyrosine 78. Among them three main types of isoquinoline derivatives are found in A. mexicana: 1- The protoberberine type includes: berberine 10, scoulerine 79 and cheilanthifoline 80. 2- The protopine type includes: allocryptopine 81, argemexicaine A 82 and B 83 and protopine 84. 3- The benzo[c]phenanthridine type includes: norchelerythrine 85, norsanguinarine 86, pancorine 87 and sanguinarine 88.

53

Figure 3. The main isoquinoline alkaloids of Argemone mexicana

54

Protoberberine alkaloids are one of the most widely distributed among the isoquinoline alkaloids groups. The classic biosynthetic route for the formation of protoberberines involves the same fundamental units as those for the formation of benzylisoquinoline alkaloids. Two molecules of tyrosine 78 are involved, one yielding dopamine 89 via DOPA, and the second proceeding to 3,4-dihydroxy-phenyl pyruvic acid 90. These molecules afford norlaudanosoline 91 by Mannich condensation followed by decarboxylation. A selective methylation of norlaudanosoline 91 provides reticuline 92, which is a precursor of many isoquinoline alkaloids (Cordell, 1981). A representative scheme of the formation of reticuline 92 from the two molecules of tyrosine 78 is shown in Figure 4.

It is well known that (+)-isomer of reticuline 92 is the true biosynthetic precursor of several alkaloids, including berberine 10. The so-called berberine bridge carbon at C-8 of the protoberberine alkaloids has been shown to be derived from the N-methyl group of methionine. The oxidation of the N- methyl group and subsequent ring closure is thought to proceed via an iminium species 93 to give either 9,10- 94 (pathway I) or 10,11- 95 substituted (pathway II) series of compounds as illustrated in Figure 5 (Cordell, 1981).

55

Figure 4. Biosynthesis of (+)-Reticuline 92, a precursor of several isoquinoline alkaloids

56

Figure 5. Formation of 9,10- or 10,11- substituted intermediates from (+)-Reticuline 92 in the biosynthesis of protoberberine alkaloids

Protoberberine alkaloids are also considered as the precursors of other skeletal structures, particularly the protopine and benzophenanthridine alkaloids. In the case of protopine alkaloids, reticuline 92 was shown to proceed to scoulerine 79 and then isocorypalmine to give allocryptopine 81. Scoulerine 79 has also been demonstrated to be an intermediate in the biosynthesis of protopine 84 via the formation of the protoberberine alkaloid cheilanthifoline 80 (Cordell, 1981).

57

2.3.1.4 Biosynthesis of benzophenanthridine alkaloids Benzophenanthridine biosynthesis also involves reticuline 92 as a common precursor. It has been suggested that these alkaloids are derived by fission of the 6,7 bond of protoberberines with subsequent bond formation between C-6 and C-13. One example is the formation of chelidonine 96 and sanguinarine 88 from reticuline 92. The latter cyclicizes into scoulerine 79 which will undergo the formation of two methylendioxy groups to produce stylopine 97. The intermediate 98, which is believed to originate from stylopinemetho salt 99, would undergo a cleavage to the enamine aldehyde 100, providing either chelidonine 96 by the reduction of the imino species 101 or sanguinarine 88 by oxidative dehydration as demonstrated in Figure 6 (Cordell, 1981).

58

Figure 6. Biosynthesis of the benzophenanthridine alkaloids Chelidonine 96 and Sanguinarine 88 from the common precursor (+)-Reticuline 92.

59

2.3.1.5 Pharmacological activity of protoberberine and protopine alkaloids From the pharmacological point of view, protoberberine and protopine alkaloids have presented a series of different in vitro and in vivo activities. Berberine 10 and its derivatives have been particularly studied and have demonstrated important antimicrobial activity, covering a range of organisms from fungi and protozoa to bacteria. Berberine sulphate shows inhibitory activity against Orynebacteriu mdiphteriae, , Xanthomonas citri and Candida tropicalis (Cordell, 1981).

Berberine 10 is able to increase membrane permeablility in bacteria and intercalate into DNA (Lewis and Ausubel, 2006) and has demonstrated a good antiprotozoal activity against several species of leishmania. In fact, this alkaloid has been used clinically for the treatment of leishmaniasis for over 50 years (Osorio et al., 2008). Among the different types of isoquinoline alkaloids, the protoberberines showed to have the most potent antiplasmodial activity against Plasmodium falciparum (strain K1). Dehydrodiscretine and berberine 10 were the most active with IC50 values lower than 1.0 µM, while the protopine-type alkaloids, protopine 84 and allocryptopine 81 had IC50 values of 34.0 and 5.1 µM, respectively (Wright et al., 2000). It has been demonstrated that berberine 10 is a potent in vitro inhibitor of both nucleic acid and protein synthesis in Plasmodium falciparum (Elford, 1986). While the mechanism of action and pharmacological activities of berberine 10 have been extensively studied, very little is known about the other protoberberine alkaloids.

60

Studies on the structure-activity relationships of protoberberine alkaloids having an antimalarial activity shows that the presence of a quaternary nitrogen in this series of alkaloids seems to increase the antimalarial activity (Osorio et al., 2008). Moreover, the activity can also be increased by the aromatization of ring C due to the quaternization of the nitrogen in ring B, and by the type of oxygen functions in rings A, C, and D (Iwasa et al., 1998; Iwasa et al., 1999; Osorio et al., 2008).

Another important feature of protoberberine and protopine alkaloids are their cardiac effects observed in vitro and in vivo. Berberine salt has been used in China to treat arrhythmia and heart failure for years. The capacity of the alkaloid to prolong the duration of cardiac action potentials is well known and this effect is mainly attributed to the inhibition of slowly activating components and increase of L-type Ca2+ currents in myocytes (Rodriguez- Menchaca et al., 2006; Wang and Zheng, 1997; Wang et al., 1996). In the same way, allocryptopine 81 has also been studied for its anti-arrhythmic properties. This alkaloid was demonstrated to prolong the refractory phase of the heart muscle, abolishing fibrillations, in the same way as quinine and quinidine (Benthe, 1956). Possible mechanisms of action for the anti- arrhythmic effect of allocryptopine 81 have been investigated showing that the alkaloid was able to produce a blocking effect on the transient outward potassium current in rabbit left-ventricular myocytes (Li et al., 2008). The alkaloid protopine 84 has been studied especially in China for its cardiac effects. An antiarrhythmic activity has also been demonstrated by the prolongation of the functional refractory period. An inhibition of spontaneous beat and contractive force were also observed (Yu et al., 1999).

61

Benzophenanthridine alkaloids present in Argemone mexicana are known to be responsible for the toxicity of Argemone oil, whose ingestion is the cause of the disease called epidemic dropsy. A great number of poisoning cases (over 3000) was reported in India during August-September 1998 due to the consumption of mustard oil adultered with Argemone seed oil. The disease is characterized by pathological accumulation of diluted lymph in body tissues and cavities (Verma et al., 2001). Sanguinarine 88 and dihydrosanguinarine are the main alkaloids in the Argemone oil, and they are present mostly in the seeds and roots of the plant. A few mechanisms have been associated with the toxicity of sanguinarine 88, such as the increase of peroxidation and changes in enzyme activity of serum in the liver. The hepatotoxicity of the alkaloid seems to be associated with oxidation of protein thiols and disturbance of mitochondrial respiration (Choy et al., 2008). It is important to notice that sanguinarine 88 has been reported as a trypanocidal agent against Trypanosoma brucei with an IC50 value of 1.9 µM. However, the activity was not specific, since the alkaloid showed high cytotoxicity (Osorio et al., 2008).

62

2.3.2 Nauclea latifolia Smith. (Rubiaceae) 2.3.2.1 The botanical description of N. latifolia Smith. N. latifolia Smith. belongs to the flowering plants family Rubiaceae, a large family of 630 genera and about 13000 species found worldwide, especially in tropical and worm regions, making the Rubiaceae one of the six largest angiosperm families including Astreaceae, Orchidaceae, Fabaceae, Poaceae and Euphorbiaceae in terms of the number of genera and species. Due to their wide distribution the plants of Rubiaceae are used in all part of the world as ornamentals, foods and remedies and continuously screened in laboratories for their pharmacological properties (Karou et al., 2011). N. latifolia is known as African quinine (Agyare et al., 2006) and African peach. It is described as a tree-plant, which is native to savannah forest of continental Africa (Abbah et al., 2010). It is scandent or straggling shrub in savannah woodland or small spreading tree, rarely over 20ft high, bole crooked, or larger tree over 100 ft high and 8 ft girth, in closed forest, rough leaves 7 by 4-5 inches, glabrous obovate. Flower-head, up to 2 diameters and shallow pitted fruits up to 3 inches in diameter, the fruits are edible, sweetly acid pulp with the numerous seed embedded (Agyare et al., 2006).

2.3.2.2 Ethnopharmacological importance of Nauclea latifolia Smith. Many reports were published described the traditional medicinal use of N. latifolia worldwide, commonly used parts of N. latifolia include the leaves, roots, stem, and fruits (Ayeleso et al., 2014). In Sierra Leone, it is used to manage venereal diseases and constipation. In Sudan, a root infusion is used to treat gonorrhoea. The bitter root or root bark is used for the management of fever and as a purgative. The roots, root bark or stem are used as chewing sticks for toothache and carries, in Sudan and southern Nigeria, the teeth of the

63 users of chewing sticks are usually clean, strong, fresh and devoid of dental plagues and carries (Agyare et al., 2006; Okwu and Uchenna, 2009). A decoction of the root bark is recommended as a mouth wash for swollen gums and the decoction of the leaves makes an efficacious gargle for swollen gums and mouth ulceration in southern Nigeria. In the north-eastern part of Nigeria, the aqueous extract of the stem bark of the plant is widely used for varied medicinal purposes including effective treatment of gastrointestinal worm infections, malaria, fever, stomach, nematodes infections and liver diseases (Onyeyili et al., 2001; Ayeleso et al., 2014). The Nigerian also used the plant in treatment of cough and gonorrhoea. The roots and leaves are used in Ghana for stomach complaint and treating sores (Agyare et al., 2006). In the , the leaves of Nauclea latifolia Smith. is used in traditional medicine for the treatment of cerebral malaria, behavioural disturbances in mentally-retarded children or central nervous system diseases, such as anxiety, depression and epilepsy (Taïwe et al., 2014).

N. latifolia has been extensively used in herbal medicine as antiviral agent for liver treatment, it protect and regenerate liver cells, viral liver damage and toxic liver damage. The fruits have broad spectrum activity against measles (Okwu and Uchenna, 2009; Abbah et al., 2010). Alkaloids and tannins in the stem bark are noted for their anti-inflammatory property and as a remedy for leucorrhoea (Okwu and Uchenna, 2009). In vitro anti-trypansomal activity of aqueous and methanolic extracts of the stem bark has been evaluated on Trypanosoma congolense. On the other hand it has also been reported that the root extract of N. latifolia have trypanocidal properties on Trypanosoma brucei. Antimalarial activity has been evaluated for N. latifolia, and alkaloids were identified as the main chemical group that

64 responsible for antimalarial activity (Maikai and kobo, 2008). The hot water extract of the root and stem bark of the plant was found to be active against Plasmodium falciparum in vitro. Further investigations revealed that the cold water extracts of the leaves of Nauclea latifolia as well as some of its fractions were active against Plasmodium berghei in vivo, in addition to their spasmolytic effects (Amos et al., 2005). The roots, leaves and stem bark extract showed antiplasmodial activity against Plasmodium vivax and Plasmodium falciparum. On the other hand a series of alkaloids comprising, nauclerofoline, nauclechine, naufoline, naucleitine, nauclefine and naucleidinal also showed antiplasmodial activity. The antioxidant potentials in the aqueous extracts of the leaves and fruits of N. latifolia have been investigated (Okwu and Uchenna, 2009; Ayeleso et al., 2014). Anti- inflammatory, antipyretic and antinociceptive activities have been evaluated for the aquous extract of the root bark of N. latifolia in mice and rats. The aqueous and ethanolic extract of N. latifolia were significantly lowered the fasting blood glucose levels of the diabetic rats. Another study on the same extracts exhibited anti-bacterial activity. The plant has been found to lowering the blood pressure in both normotensive and hypertensive rats (Nworgu et al., 2008; Karou et al., 2011; Ayeleso et al., 2014). Antinociceptive effects of the alkaloids fraction isolated from N. latifolia have been evaluated (Taïwe et al., 2014).

Previous scientific studies of the plant revealed that both the methanol and ethanol extracts of the dried fruits, stem and root bark possess spasmolytic and anti-bacterial activity. The cardiovascular effect of the roots and leaves extracts of N. latifolia have also been reported. The neuropharmacological effects of the aqueous extract of the root bark in rodents have been evaluated

65

(Amos et al., 2005; Abbah et al., 2010). The fruits extract was shown to be active against Human Immune deficiency Virus (HIV) (Hussein et al., 1999; Nworgu et al., 2008).

2.3.2.3 Phytochemistry of Nauclea latifolia Smith. Previous phytochemical investigation of the plant revealed the presence of alkaloids such as, indole quinolizidine alkaloids (glycol alkaloids), flavonoids, tannins, saponins, anthraquinones, steroids, glycosides and phenolic compounds. The fruits contain copper, iron, cobalt, calcium, magnesium, zinc, phosphorous and vitamins (A, B1, B2, C, and E) (Okwu and Uchenna, 2009; Ayeleso et al., 2014; Taïwe et al., 2014). The presence of monoterpene, triterpene, indole alkaloids, saponins and traces of inorganic compounds in the roots has also been reported (Abbah et al., 2010). The indole alkaloid nauclefolinine have been isolated from the roots of N. latifolia (Ngnokam et al., 2003).

2.3.2.4 Biosynthesis of monoterpene indole alkaloid Strictosamide 44 More than 3000 terpenoid indole alkaloids are recognized, making this one of the major groups of alkaloids in plants. They are found mainly in eight plant families, of which the Apocynaceae, the Loganiaceae, and the Rubiaceae provide the best sources. In terms of structural complexity, many of these alkaloids are quite outstanding and it is attributed to the painstaking experimental studies of various groups of workers that we are able to rationalize these structures in terms of their biochemical origins. In virtually all structures, a tryptamine 102 portion can be recognized. The remaining fragment is usually a C9 or C10 residue, and three main structural types are discernible. The C9 or C10 fragment was shown to be of terpenoid

66 origin and the secoiridoid, secologanin 103 was identified as the terpenoid derivative, which initially combined with the tryptamine 102 portion of the molecule. A large number of iridoids are found as glycosides, e.g. loganin 104, glycosylation effectively transforming the loganin 104 is a key intermediate in the biosynthesis of many other iridoid structures, and also features in the pathway to a range of complex terpenoid indole alkaloids. Fundamental in this further metabolism is cleavage of the simple monoterpene skeleton still recognizable in loganin 104 to give secologanin 103, representative of the secoiridoids. Secologanin 103 now contains a free aldehyde group, together with further aldehyde and enol groups, these latter two fixed as an acetal by the presence of the glucose.

Figure 7. Biosynthesis pathway of monoterpene indole alkaloid Strictosamide 44 TDC: Tryptophan decarboxylase; G10H: Geraniol 10 hydroxylase; SLS: Secologanin synthase; STR: Strictosidine synthase

67

2.3.2.5 Pharmacological activity of β-carboline alkaloids The genus Nauclea (Rubiaceae) is a rich source of alkaloids. To date, more than 40 monoterpenoid indole alkaloids have been isolated from this genus. Some of these alkaloids were reported to have antiproliferative, antiparasitic, and antimicrobial activities (Zhang et al., 2001). A number of monoterpene indole alkaloids of β-carboline type such as nauclefine and naulafine have been isolated from plants of Nauclea species. Recently, several monoterpene indole alkaloids have been isolated from Nauclea orientalis, which have interesting biological activity. N. orientalis L. is a tree found in Papua New Guinea, Indonesia, Peru and Queensland (Australia). Villagers of the Central Province of Papua New Guinea use the bark of this tree to treat abdominal pain, animal bites and wounds, while its leaves are used traditionally by coastal Australian Aborigines as a pain-killer and fish poison. It have been reported that several indole alkaloids have been isolated from the ammonical and ethyl acetate extracts of N. orientalis leaves. The monoterpene indole alkaloids, strictosamide 44 and vincosamide 105 were isolated from the bark of N. orientalis. Monoterpene indole alkaloids, naucleamides A—E (A 106; B 107; E 108) have been isolated from the bark and wood of Nauclea latifolia (Shigemori et al., 2003; Zhang et al., 2001). The chloroform and butanol fractions from the ethanol extract of the bark of N. orientalis were found to be active in a screening assay for inhibition of the secreted aspartic protease (SAP) of Candida albicans from higher plants, SAP have been shown to be a major virulence factor in Candida infections. Inhibition of SAP has been proposed as a new approach in the treatment of candidosis. The phytochemical screening of these fractions revealed the isolation of the indole alkaloids,

68 nauclealines A 109 and B 110 and naucleosides A 111 and B 112 as well as strictosamide 44 and vincosamide 105 (Zhang et al., 2001).

Figure 8. Chemical structures of indole alkaloids from Nauclea spp. that exhibited many biological activities.

69

3. MATERIALS AND METHODS

3.1 General experimental conditions 3.1.1 Organic solvents, glassware, chemicals and reagents -Organic solvents used in the extraction and fractionation Commercial solvents used in the extraction and fractionation included n- hexane, petroleum ether, chloroform, dichloromethane, ethyl acetate, acetone, ethanol and methanol after their distillation. - HPLC grade methanol, acetonitrile and water were used in the elution. - Glassware The glassware used included, TLC developing tanks, glass columns of various sizes, conical flasks, round button flasks, beakers, , vials, measuring cylinders, funnels, separating funnels, , petri dishes, porcelain dishes and mortars.

3.1.2 Spectroscopic techniques

UV spectra were presented as λmax nm (log ԑ) and recorded on a Shimadzu UV240 spectrophotometer in methanol solution. IR spectra were presented in cm-1 and recorded on KBR discs on a JASCO A- 302 spectrophotometer. 1HNMR and 13CNMR spectra were recorded on AV-600, AV-500, AV-400 and AV-300.13CNMR were recorded at 150,125, 100 and 75 MHz. 2D-NMR spectra were recorded on Bruker AV-500 NMR spectrometer. The chemical sifts values were presented in δ (ppm), the solvent proton signals of CD3OD and CDCl3 were taken as reference. The coupling constant value (J) was calculated in Hz. Low resolution electron-impact mass (EI/MS) spectra, chemical ionization (CI/MS) and gas chromatography (GC/MS) were recorded on JMS-600 H

70

Jeol, Japan. High resolution EI/MS were recorded on MAT 95-XP (Thermo- Finnigan, Germany). Low and high resolution fast atom bombardment mass spectra (FAB/MS) were recorded on HX-110 Jeol, Japan.

Liquid chromatography mass spectra (LC/MS) and electro spray ionization (ESI/MS) high resolution were recorded on Applied Bio-system QSTAR® XL LC/MS/MS system, USA and measured in a glycerol matrix. MALDI/TOF/TOF was recorded on BRUKER ultraflex 111 TOF/TOF.

3.1.3 Chromatography Thin layer chromatography (TLC) was performed on pre-coated silica gel ® plates (ALUGRAM SIL 60 G/UV254 - DC (E. Merck, 20X20X0.5 mm thick).

Column chromatography were performed on silica gel (E. Merck, 40-60 and 200-400 μm mesh size), ODS C-18 (63-212 μm, Japan) Diaion HP-20 (Mitsubishi, chem. Ind., Tokyo, Japan), sephadex (LH-20) and alumina.

Recycling preparative high performance liquid chromatography (size exclusion) (RPHPLC) was performed on (Jaigel LC-908W - C 60, Japan analytical industry, Co, Ltd.) equipped with YMC ODS L-80 or H-80 and a column GS-320 (YMC Ltd. Japan), the flow rate was 4 mL/minute, the solvent system used was methanol. Reverse phase HPLC was performed on Jaigel column 00S L-80, LC-908, the flow rate was 4 mL/ minute; the solvent system used was methanol/water. Analytical HPLC was performed on WATERS 2695, the solvent system used was methanol/water., the flow rate was 1.0 mL/minute, column B260B, OD-5-100/25; C-18 and 4.6X 250 mm size.

71

3.1.4 Detection method on TLC by spraying reagents Initially the spots were visualized under UV light (254 and 366 nm), then sprayed with ceric (IV) sulphate or Vanillin/sulphuric acid and heated till the appearance of the coloured spots. The spots were detected also by iodine vapor by inserting the TLC plate in a tank containing a mixture of few iodine crystals and silica gel, till the appearance of the spots. For detection of alkaloids Dragendorff’s reagent was used.

3.2 Plant Materials Two Sudanese medicinal plants namely, Argemone mexicana L. (aerial parts) and Nauclea latifolia Smith. (root bark), belong to the families Papaveraceae and Rubiaceae, respectively, were collected and authenticated and herbarium specimens were deposited in the Faculty of Pharmacy, University of Science & Technology, Omdurman, Sudan.

3.2.1 Extraction and fractionation of plant materials Plant materials were collected and shade dried, then crushed in a mechanical grinder to obtain coarse powder. The pulverized materials (100 g of each plant part) were successively extracted by 70% ethanol for five hours in a water bath. The ethanolic crude extracts were concentrated and the remaining aqueous portion was fractionated sequentially by petroleum ether (bp. 60- 80°C), chloroform and ethyl acetate. Each of the organic solvent fractions and the water resdue were further concentrated under reduced pressure in a Büchi Rotavapor R-200.

72

3.3 Phytochemical screening Each of the crude extracts and their respective fractions were subjected to phytochemical screening on TLC using pre-coated silica gel and/or alumina plate to detect the number and nature of compounds present using different mobile solvent systems. The dried TLC plate were visualized under UV and sprayed with chromogenic reagents. This preliminary analysis also served to select the optimum suitable mobile solvent system for atmospheric pressure column chromatography, High Performance Liquid Chromatography (HPLC) and/or preparative TLC. Finally the purified isolated compounds were subjected to spectroscopic techniques, UV, IR, NMR and Mass spectrometry for identification and structure elucidation.

3.3.1 Isolation of compounds from Argemone mexicana L.

3.3.1.1 Isolation of compounds from petroleum ether fraction The chromatographic process was commenced with loading the petroleum ether fraction (1.7365 g) into a silica gel column (20Х3 cm). The eluent was initially n-hexane and polarity was increased gradually by adding DCM followed by methanol and finally the column was washed by pure methanol. A number of sub-fractions were obtained; on further repetitive column chromatography for sub-fraction no. 4 (1.0 g) over silica gel column (22X2 cm) using the same mobile solvent system a number of 250 sub-fractions were obtained. The sub-fractions (61–80) were pooled together and on TLC showed two spots after development in a mobile phase composed of 30% hexane/DCM. The two compounds were further purified by preparative silica gel TLC using the same mobile system, to eventually yield 3.0 mg of a colourless amorphous compound, soluble in DCM which was designated as

73

(AM/1) 113 (Scheme 1)., The compound gave negative reaction with Dragendorff’s reagent. On the other hand, the sub-fractions (136–155) were combined and subjected to preparative silica gel TLC using 20% hexane/DCM as a mobile phase. The eluted band after repeated recrystallization yielded faint brownish fine crystals (6.0 mg) soluble in DCM, which gave a positive reaction with Dragendorff’s reagent. This compound was designated as (AM/2) 85 (Scheme 1).

3.3.1.2 Isolation of compounds from ethyl acetate fraction The ethyl acetate fraction (2.5 g) was loaded in a silica gel column (18.5X3 cm) and was eluted with a mixture of DCM/ethyl acetate and the , polarity was consistently increased till washing the column by 100% methanol. A number of 95 sub-fractions were obtained. The sub-fractions (89–91) were pooled together due to their chromatographic similarity on silica gel TLC using 95% DCM/methanol as a mobile phase. On further purification by preparative silica gel TLC a highly fluorescent compound was eluted to yield a phosphorus amorphous compound (20 mg) which was positively reacted with Dragendorff’s reagent and soluble in methanol. This compound was designated as (AM/3) 10 (Scheme 1). The chloroform fraction yielded non compounds.

74

Scheme 1. Isolation of the compounds (AM/1), (AM/2) and (AM/3) from Argemone mexicana L. (aerial parts).

75

3.3.2 Isolation of compounds from Nauclea latifolia Smith. 3.3.2.1 Isolation of compounds from petroleum ether fraction The petroleum ether fraction (0.8476 g) obtained from the N. latifolia crude extract was subjected to column chromatography over silica gel (26X2.5 cm) and eluted with hexane/acetone and finally washed by pure methanol. A number of 32 sub-fractions were obtained; sub-fractions 7 and 8 were combined together (0.1988 g) and further subjected to a sephadex column (LH-20) (55.5X2.5 cm) using DCM/methanol (1:1) as a mobile phase. A total of 58 sub-fractions were obtained, among them (9-25) were pooled together and resulted in the formation of colourless crystals which on recrystallization in methanol yielded (3.5 mg) of compound reacted positively with ceric (IV) sulphate as spray reagent and gave a negative results when sprayed with Dragendorff’s reagent. This compound was designated as (NL/1) 114 (Scheme 2).

3.3.2.2 Isolation of compounds from chloroform fraction The chloroform fraction (0.6696 g) was column chromatographed over silica gel (23.5X2.5 cm), using hexane/acetone as eluent. This column provided 122 sub-fractions. Sub-fractions (15-30) were pooled together due to their chromatographic similarity and on concentration they yielded (0.0194 g) of dray material which was further subjected to a sephadex column (LH-20) (55X2.5 cm) and eluted with pure methanol. From the 41 sub-fractions obtained, similar sub-fractions (20-31) were combined together on the basis of their TLC evidence on silica gel using 70% hexane/acetone as a mobile phase. This resulted in a pure compound (1.2 mg) which reacted positively on sprying with Dragendorff’s on silica gel TLC. This compound was designated as (NL/2) 115 (Scheme 2).

76

Scheme 2. Isolatation of the compounds (NL/1), (NL/2) and (NL/3) from Nauclea latifolia Smith. (Root bark).

77

3.3.2.3 Isolation of compounds from ethyl acetate fraction The preliminary analysis of the ethyl acetate fraction by TLC indicated the presence of alkaloids; hence Dragendorff’s reagent gave clear major orange spot. The ethyl acetate fraction (0.9954 g) was subjected to alumina column (30X2.5 cm) and eluted with a mixture of hexane/acetone after adding two drops of methyl amine. A number of 54 sub-fractions were obtained. On Further purification of the pooled and concentrated sub-fractions 29-43 (461.8 mg) by reverse phase HPLC (80% methanol/water) yielded (300 mg) of compound designated as (NL/3) 44 (Scheme 2).

3.4 Biological assays 3.4.1 Materials MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide) was obtained from Acros organics, (NJ 07410, USA); Dulbecco's Modified Eagle's Medium – high glucose (DMEM), and minimum essential medium eagle (MEM), were from sigma Aldrich, (St. Louis, MO, USA); RPMI-1640 was from Mediatech Inc. (Herndon, VA, USA); Triton x-100 from MERK Millipore, (Darmstadt, Deutschland, Germany); and dimethyl sulfoxide (DMSO) was from Fisher Scientific, (Loughborough, UK); HEPES free acid from MP Biomedicals, (67402 Illkirch, France); Trypsin-EDTA from Gibco, (Grand Island, NY 14072. USA).

78

3.4.2 Antileishmanial assay In this study the plants extracts, fractions and the pure compounds isolated from them were screened in vitro against the extracellular, promastigote and intracellular, amastigote forms of three leishmania species, L. major, L.tropica and L. donovani. The assay against the promastigote involved colorimetric method, using ELIZA reader at 490 and 570 nm, and the assay against the amastigote involved the macrophage method, in which macrophage cells were taken from albino mice and 96 well plates were used for the assay. Amphotericin B and pentamidine were used as reference drugs (positive controls) against the promastigote form of parasite in the colorimetric method, while sodium stibogloconate (pentostam®) was used against the amastigote in the macrophage cells method. The counting chamber was used for counting the cells and parasites and observed under the inverted microscope. Incubators and centrifuge were also used.

3.4.2.1 Maintenance of Parasites 3.4.2.1.1 Preparation of Biphasic medium (NNN medium) The blood was taken from rabbit in sterile condition, then Gentamicine was added to the blood and poured the blood in agar (melted agar), autoclaved, and cooled (45-50°C), then dispense in autoclaved screw cupped vials and kept in slanted position.

3.4.2.1.2 Preparation of complete RPMI 1640 medium Fetal calf serum (FCS 10%) and antibiotic (Gentamicine) were added to RPMI 1640. In sterile conditions and kept on fridge.

79

3.4.2.1.3 Subculture of parasites The parasites were sub cultured in a new biphasic media bottles after the addition of complete RPMI and incubated in cooled incubator (23°C). 3.4.2.2 Preparation of sample An amount of 50 μL of DMSO and 95 μL complete RPMI 1640 were added to 1 mg of sample (extract, fraction or pure compound).

3.4.2.3 In vitro assay by colorimetric method 3.4.2.3.1 In vitro assay against extracellular promastigote Three 96 well plates were used for the three parasite strains. The amount of 180 μL of RPMI 1640 was added to the first row of each plate, 100 μL was added to all wells. 20 μL of test sample was added in first row, then applied 2- fold serially dilution method, after that 100 μL of parasites promastigote (1 x 106 cells/mL) were added in each well. Two rows were left for positive and negative control. Positive control contains antibiotics as Amphotericin B and Pentamidine, while negative control contains only media and parasites. The plate was incubated for 72 hours at 24°C. After 72 hrs, 10 μL of MTT dye was added to each well, then again the plate was incubated at 37°C for 4 hours, after four hours, the plate was centrifuged at 2000 rpm for 5 minutes, then the supernatant was discarded and the pellet was dissolved by 200 μL DMSO, finally the absorbance was taken by ELIZA reader at 490 and 570 nm (Choudhary et al, 2005; Habtemariam, 2003).

80

3.4.2.4 In vitro assay by macrophage method 3.4.2.4.1 Procedure for harvesting macrophages from the mice

2 mL simple RPMI-1640 and 1 mL of sodium citrate (anticlotting agent) were used in this study. Mice were killed by cervical fracture. The skin over the abdominal region was removed and 3 mL of the washing fluid was forced into the peritoneum with a hypodermic needle. After light massage for a few seconds, fluid was withdrawn from the lateral part of the peritoneal cavity. Usually about 2 mL fluid was obtained from each mouse, 5 mL incomplete RPMI medium were inoculated in tissue culture flask containing mouse peritoneal fluid and incubate at 37ºC for 24 hrs. Non-adherent cells were removed and macrophages were further incubated overnight in RPMI 1640 medium supplemented with 10% fetal calf serum.

3.4.2.4.2 In vitro assay against intracellular amastigotes Adherent cells were infected with promastigotes at a parasite/macrophage ratio of 6:1 and incubated for 1 hr at 37ºC in 5% CO2. Next, free promastigotes were removed by extensive washing with PBS (PH, 7.2). After 24 hrs, infected macrophages were treated at different concentrations of extracts and fractions. After 72 hrs monolayer were washed with PBS at 37ºC, fixed in methanol and stained with Giemsa. The macrophages infected with promastigotes were treated with 0.5% DMSO and used as negative control. Inhibition of parasite (%PI) was calculated by the following equation:

%PI is used further for calculation of the IC50 by using MS excel software

81

3.4.3 Cytotoxicity assay

3.4.3.1 Cell-line In vitro cytotoxicity assays were performed as described by Scudiero et al., 1988, using the 3T3 NIH mouse embryo fibroblast cell line and MDBK, a cow kidney epithelial cell line from American Type Culture Collection ‘ATCC’, (Manassas, VA 20108, USA), and CC-1, a rat Wistar hepatocyte cell line from European Collection of Cell Cultures, (Salisbury, UK).

3.4.3.2 Method The CC-1 cells were cultured in MEM supplemented with 10% FBS, 2 mM glutamine, and 20 mM HEPES. While the 3T3 cells were maintained in DMEM formulated with 10% FBS and the MDBK cell lines were grown in RPMI supplemented with 10% FBS and 2 mM L-glutamine. All of these cells are adherent cells and required to be detached from culture flasks surfaces by trypsin/EDTA treatment. The media were removed from the cell culture and sterile PBS was added to each flask to wash cells from cell debris and aspirated out. Then, 0.25% Trypsin/EDTA solution was added to the attached cells and incubated for 3 min at 37C after which, flasks were gently tapped and observed under microscope to check for detachment of cells from flask surfaces followed by addition of media containing FBS. Cells were collected in 15 mL centrifuge tubes and centrifuged at 1200 rpm. The pellet was resuspended in complete media and cells were counted using microscope and Neubauer counting chamber.

82

The MTT assays for the 3T3 and CC-1 cells were performed by using 6 103 cell/well in 100 µL complete media in a flat-bottomed 96 well plate, while that for MDBK cells was done by using 1  104 cells per well/100 µL in the complete media. All plates were incubated for 24 hrs at 37C in CO2 incubator. After attachment of cells, media was replaced by 200 µl media containing compounds at different concentrations (1, 5, and 20 µg/mL) in triplicates and again incubated for 48 hrs at 37C in CO2 incubator. Following exposure to each compound, cell viability was assessed by using 0.5 mg/mL of MTT in complete media for 4 hrs followed by the removal of supernatant and addition of 100 µL of DMSO to each well to solubilize the formazan complex formed by the action of mitochondrial dehydrogenases. The plates were read at 540 nm after one minute shaking and optical density readings were processed using MS Excel software. The results were expressed as means ± SD of triplicate readings.

83

4. RESULTS AND DISCUSSION

4.1 Secondary metabolites isolated from the aerial parts of Argemone mexicana L. The concentrated petroleum ether fraction was column chromatographed over silica gel, and eluted with a mixture of hexane/DCM. The eluted sub-fraction (4) was subjected to another silica gel column using the same solvent system. The sub fractions (61- 80) were pooled and showed two major spots on TLC, which yielded compounds AM/1 on further purification by preparative TLC.. On the other hand the sub fractions (136–155) were combined and subjected to preparative silica gel TLC, The eluted band after repeated recrystallization yielded AM/2 as depicted in Scheme 1.

The concentrated ethyl acetate fraction was subjected to column chromatography over silica gel. The solvent system hexane/DCM is used for elution. On further purification of the combined sub-fractions (89 – 91) by preparative TLC a highly fluorescent yellow compound was obtained and designated as AM/3 (Scheme 1).

4.1.1 Characterization of AM/1 as Sitost-4-en-3-one (β-Sitostenone) 113 AM/1 was obtained as fine colorless needle from petroleum ether fraction.

TLC on silica gel F245 showed a single dark spot under UV light (254 nm), which reacted negatively with Dragendorff’s reagent. EI/MS showed molecular ion peak (M+) at m/z 412 corresponding to the molecular formula + C29H48O established by HREI/MS. FAB/MS (+ve) showed peak (M+H) at 413, this was confirmed further by ESI/MS (+ve) which showed very sharp peak (M+H)+ at 413.

84

1HNMR spectrum showed signals at δ 0.69 (3H, s), δ 0.80 (3H, d), δ 0.82 (3H, d), δ 0.91 (3H, d) and δ 1.16 (3H, s). The singlet at δ 5.70 can be safely assigned to the deshielded proton on C-6 which is affected by the carbonyl group. This data agrees with the published data for β- sitostenone 113, δ 0.71 (3H, s), 0.80 (3H, d, J = 6.4 Hz), 0.83 (3H, d, J= 6.8 Hz), 0.84 (3H, t, J= 6.8 Hz), 0.91 (3H, d, J= 6.4 Hz), δ 1.18 (3H, s) and δ 5.72 (1H, s, H-6). 13CNMR and DEPT spectra indicated the presence of eleven methylene groups and nine CH groups. The down field carbon at δ 199.7 indicated the presence of a carbonyl group. All 13C values were in agreement with the literature data reported for β- sitostenone 113 as illustrated in Table 3 (Kan et al., 2011). β-sitostenone 113 is isolated for the first time from Argemone mexicana and also from Papaveraceae family, it is considered as a new source (DNP database, 2011). Antileishmanial activity for this compound was not reported.

Structure of β-Sitostenone 113

85

1 13 Table 3. HNMR and CNMR spectral data for compound AM/1 113, (CDCl3, 1H: 500 MHz, 13C:125 MHz δ (ppm)) Compared with the literature data 1 13 (CDCl3, H: 400 MHz, C:100 MHz) (Kan et al., 2011) 1H 13C H (multiplicity, J in Hz) C Position Experimental Reference Experimental Reference (CDCl3) (CDCl3) (CDCl3) (CDCl3) 1 35.8 35.7 2 33.9 34.0 3 199.7 199.6 4 123.7 123.7 5 171.8 171.6 6 5.70 (s,1H) 5.72 (s,1H) 32.9 33.0 7 32.0 32.1 8 35.6 35.7 9 53.7 53.8 10 38.7 38.6 11 19.8 19.9 12 39.5 39.6 13 42.3 42.4 14 55.9 55.9 15 24.1 24.2 16 28.1 28.2 17 56.0 56.1 18 11.9 11.9 19 17.3 17.4 20 36.1 36.1 21 20.1 20.0 22 33.8 33.9 23 25.9 26.1 24 45.7 45.9 25 29.0 29.2 26 21.0 21.1 27 18.9 19.0 28 23.0 23.0 29 11.9 12.0

86

4.1.2 Characterization of AM/2 as norchelerythrine 85 AM/2 appeared as faint brown crystals and after further purification yielded colourless crystals with a melting point of 210 – 213°C. TLC plate showed a single blue fluorescent spot under UV light (254 – 366 nm), which reacted positively with Dragendorff’s reagent. EI/MS showed molecular ion peak (M+) at m/z 333. High resolution EI/MS established the molecular formula

C20H15NO4. ESI/MS (+ve) and MALDI/TOF/TOF showed a sharp peak (M+H)+ at m/z 334 which substantiated the EI/MS results. 1HNMR spectrum of AM/2 (Table 4) exhibited two singlets (3H each) at δ 4.04 and δ 4.10 indicating the presence of two methoxy groups, whiles the two protons singlet at δ 6.11 could be assigned to a methylenedioxy group. The aromatic region of the spectrum integrated for six protons, two singlet at δ 9.7 and δ 8.7 for C-6 and C-4 protons respectively, two doublets at δ 7.57 and δ 7.84 for C-9 and C-10 protons respectively and the doublet at δ 8.35 for the two protons on C-11 and C-12. The peak for a highly downfield shifted proton at δ 9.7 at C-6 is consistent with amine group. This evidence was further substantiated by 13CNMR hence the carbon associated with this proton appeared at δ 145.1. 13CNMR spectrum revealed presence of two methoxy groups at δ 61.9 and 56.7 peaks for C-7 and C-8, respectively. The carbon at δ 101.3 could be assigned to a methylenedioxy group. Both 1HNMR and 13CNMR spectral data of AM/2 (Table 4) are in full agreement with the published data for norchelerythrine 85 (Martin et al., 2005). Norchelerythrine 85 was previously reported from Argemone mexicana and from other species of the families Papaveraceae and Rutaceae (DNP database, 2011).

87

Structure of norchelerythrine 85 1 13 Table 4. HNMR and CNMR spectral data for compound AM/2 85, (CDCl3, 1H: 500 MHz, 13C:125 MHz δ (ppm)) Compared with the literature data (DMSO, 1H: 400 MHz, 13C:100 MHz) (Martin et al., 2005) 1H 13C H (multiplicity, J in Hz) C Position Experimenta Reference Experimental Reference l (DMSO) (CDCl3) (DMSO) (CDCl3) 1 8.7 (s, 1H) - 104.3 104.4 2 - - 148.4 148.2 3 - - 148.2 148.2 4 7.23 (s, 1H) 8.6 ( s, 1H) 102.1 100.7 4a - - 121.8 126.9 4b - - 139.9 136.9 6 9.73 (s, 1H) 9.6 (s, 1H) 145.1 145.5 6a - - 119.9 120.6 7 - - 146.5 144.1 8 - - 149.3 149.4 9 7.57 (d, 1H) 7.88 (d, (9.0)1H) 118.3 120.5 10 7.84 (d, 1H) 8.61 (d, (9.0)1H) 118.6 118.6 10a - - 129.7 127.3 10b - - 129.1 120.0 11 - 8.58 (d, (9.0)1H) 118.2 118.4 12 8.35(dd, 2H) 8.00 (d, (9.0)1H) 127.0 127.6 12a - - 128.0 129.5 O-CH2-O 6.11 (s, 2H) 6.23 ( s, 2H) 101.3 101.4 C7-OMe 4.04 (s, 3H) 4.06 ( s, 3H) 61.9 61.5 C8-OMe 4.10 (s, 3H) 4.03 (s, 3H) 56.7 56.7

88

4.1.3 Characterization of AM/3 as berberine 10 AM/3 is collected as a yellow amorphous powder. The TLC plate showed a single yellow spot under day light and yellow fluorescent under UV light (254 & 366 nm) which reacted positively with Dragendorff’s reagent. ESI/MS showed a peak (M+) at m/z 336 corresponding to the molecular formula + 1 C20H18NO4 . HNMR spectrum of AM/3 (Table 5) showed two protons singlet at δ 6.10, which could be assigned to a methylenedioxy group. The two singlets (3H each) at δ 4.19 and δ 4.10 are indicative of the presence of two methoxy groups. The aromatic region of the spectrum integrated for six protons, two singlets at δ 8.71 and δ 8.53 for C-6 and C-4 protons, respectively, and two doublets at δ 8.12 and δ 8.00 for C-11 and C-12 protons, 13 respectively. CNMR spectrum indicated the presence of two CH3 at δ 62.52 and δ 57.62 which could be assigned for the two methoxy groups at C-9 and C-10, respectively. The spectrum showed three methylene groups, one at δ 103.70 representing a methylenedioxy group. The rest of signals were in full agreement with the reported values for the quaternary protoberberine alkaloid, berberine 10 (Leyva-Peralta et al, 2015). Berberine 10 was previously reported from Argemone mexicana, Berberis and Mahonia spp. (Berberidaceae) and from many other spp. in several different families (DNP database, 2011). The antileishmanial activity of berberine 10 against Leishmania brazliensis (panamensis) and Leishmania donovani was previously reported (Vennerstrom et al., 1990; Saha et al., 2011).

89

Structure of berberine 10

Table 5. 1H and 13CNMR spectral data for compound AM/3 10, (MeOD, 1H: 500 MHz, 13C:125 MHz δ (ppm)) Compared with the literature data (DMSO, 1H: 400 MHz, 13C:100 MHz) (Leyva-Peralta et al., 2015)

1H 13C H (multiplicity, J in Hz) C Position Experimental Reference Experimental Reference (CDCl3) (DMSO) (CDCl3) (DMSO) 1 8.7 (s, 1H) - 104.3 104.4 2 - - 148.4 148.2 3 - - 148.2 148.2 4 7.23 (s, 1H) 8.6 (s, 1H) 102.1 100.7 4a - - 121.8 126.9 4b - - 139.9 136.9 6 9.73 (s, 1H) 9.6 (s, 1H) 145.1 145.5 6a - - 119.9 120.6 7 - - 146.5 144.1 8 - - 149.3 149.4 9 7.57 (d, 1H) 7.88, d (9.0)1H 118.3 120.5 10 7.84 (d, 1H) 8.61(d, (9.0)1H 118.6 118.6 10a - - 129.7 127.3 10b - - 129.1 120.0 11 - 8.12 (d, 1H) 118.2 118.4 12 - 8.00 (d, 1H) 127.0 127.6 12a - - 128.0 129.5 O-CH2-O 6.10 (s, 2H) (s, 2H) 103.7 103.6 C9-OMe 4.19 (s, 3H) (s, 3H) 62.5 62.4 C10OMe 4.10 (s, 3H) (s, 3H) 57.6 57.5

90

4.2 Secondary metabolites isolated from the root bark of Nauclea latifolia Smith. The petroleum ether fraction was column chromatographed over silica gel, and eluted with hexane/acetone. The sub-fractions (7 & 8) eluted from the silica column was combined and further chromatographed on sephadex column (LH- 20) using the solvent system DCM/methanol (1:1) for elution to eventually yield a colourless crystals designated as NL/1. The chloroform fraction was loaded on top of a silica gel column and chromatographed by elution with hexane/acetone. Repetitive column chromatography over sephadex (LH-20) using methanol as eluent for the sub- fractions (15-30) and pooling the chromatographically identical sub-fractions obtained (21-30) yielded a greenish – yellow amorphous compound designated as NL/2. The ethyl acetate fraction was column chromatographed over alumina column and eluted with a mixture of hexane/acetone and two drops of methyl amine. Further purification of the eluted sub-fractions (29 – 43) by reverse phase HPLC (80% methanol/water) yielded compound NL/3.

4.2.1 Characterization of NL/1 as β-Sitosterol 114 NL/1 was obtained as a white amorphous and exhibited optical activity with

[α] -55.6º (c 0.09, CHCl3) solid.TLC plate showed a single red spot when sprayed with ceric (IV) sulphate; the spot was turn to black after some time and reacted negatively with Dragendorff’s reagent. UV spectrum showed absorption maxima at 203 nm (Kolak et al., 2005). IR spectrum, vibration at 3425 and 2925 cm−1 were observed, indicating OH and sp3 C-H stretching vibration respectively. The other peaks at 1465 and 1375 cm−1 were due to

CH2 and CH3 bending.

91

EI/MS showed molecular ion peak (M+) at m/z 414 with a fragments at 396

(-H2O), 381, 273, 255 and 213, these values were identical to the EI/MS fragments reported for β-sitosterol 114 (Rao et al., 2011). High resolution mass showed the molecular formula C29H50O corresponding to the accurate mass 414. The ESI/MS showed a pseudomolecular ion peak [M+H]+ at m/z 415. The 1HNMR spectrum (Table 6) showed a broad singlet at downfield region of δH 5.34 which attributed to H-6 indicative of the presence of double bond functionality between C-5 and C-6. Two singlet of methyl groups were observed at δH 0.67 and δH 0.99 which attributed to 3H-18 and 3H-19 respectively. Two methyl protons of 3H-21 and 3H-29 were observed as multiplets at δH 0.89 – 0.91 and δH 0.84 – 0.89, respectively, while the another two methyl protons; 3H-26 and 3H-27 were overlapped at δH 0.78 0.83. The 13CNMR and DEPT spectra (Table 6) of β-sitosterol 114 indicated a total of twenty nine carbon signals; six methyl, eleven methylenes, nine methines and three quaternary carbons which was in agreement with the molecular formula of β-sitosterol 114. The carbon signal at δC 71.8 indicated the presence of oxymethine proton at C-3. All 13CNMR values (Table 6) were identical with the reported values for β-sitosterol 114 (Kolak et al., 2005; Rao et al., 2011). Complete 1HNMR and 13CNMR assignments (Table 6) were established by analysis of 2D-NMR data as well as reported data, thus the compound was identified as β-sitosterol 114. β-sitosterol 114 is isolated for the first time from Nauclea latifolia and Rubiaceae family (DNP database, 2011). Antileishmanial activity of β-Sitosterol 114 against Leishmania major was reported before (Iranshahi et al., 2007).

92

1 13 Table 6. HNMR and CNMR spectral data for compound NL/1 114, (CDCl3, 1H: 500 MHz, 13C:125 MHz δ (ppm)) compared with the literature data 1 13 (CDCl3, H: 400 MHz, C:100 MHz) (Kolak et al., 2005; Rao et al., 2011) 1H 13C H(multiplicity, J in Hz) C Position Experimental Reference Experimental Reference (CDCl3) (CDCl3) (CDCl3) (CDCl3) 1a 1.81-1.85 (m) - 37.3 37.3 1b 1.03-1.08 (m) - - - 2a 1.94-2.02 (m) - 32.0 31.6 2b 1.42-1.63 (m) - - - 3 3.48-3.55 (m) 3.52 (m) 71.8 71.8 4 2.18-2.27 (m) - 42.3 42.2 5 - - 140.8 140.8 6 5.34 (br s) 5.36 (br s) 121.8 121.7 7a 1.94-2.02 (m) - 32.0 31.9 7b 1.42-1.63 (m) - - 8 1.81-1.85 (m) - 31.7 31.9 9 0.89-0.91 (m) - 50.2 51.2 10 - - 36.6 36.5 11 1.42-1.63 (m) - 21.2 21.1 12a 1.94-2.02 (m) - 39.8 39.8 12b 1.03-1.18 (m) - - - 13 - - 42.4 42.3 14 0.99 (s) - 56.8 56.8 15 1.03-1.18 (m) - 24.8 24.3 16 1.81-1.85 (m) - 28.3 28.3 17 1.03-1.18 (m) - 56.1 56.0 18 0.67 (s) 0.68 (s) 11.9 11.9 19 0.98 (s) 1.01 (s) 19.5 19.4 20 1.24-1.38 (m) - 36.2 36.2 21 0.89-0.91 (m) 0.92 (d, 6.4) 18.9 18.8 22a 2.30-2.35 (m) - 33.8 33.9 22b 0.94-0.98 (m) - - - 23 1.03-1.18 (m) - 26.1 26.1 24 0.89-0.91 (m) - 45.9 45.9 25 1.59-1.63 (m) - 29.2 29.2 26 0.78-0.83 (m) 0.81 (d, 6.5) 19.9 19.8 27 0.78-0.83 (m) 0.83 (d, 6.5) 19.1 19.3 28 1.24-1.38 (m) - 23.1 23.1 29 0.84-0.86 (m) 0.85 (t, 7.5) 12.1 12.2

93

Structure of β-Sitosterol 114

4.2.2 Characterization of NL/2 as naucleficine 115 NL/2 exhibited a highly fluorescent one single spot on TLC when visualized under UV (366 nm). The compound gave orange spot when sprayed with Dragendorff’s reagent. UV spectrum showed clear characteristic peaks for indole alkaloids at 192, 292, 366 and maxima at 206 nm, these values were in agreement with the reported values for naucleficine 115. EI/MS showed molecular ion peak (M+) at m/z 314 which supported the molecular formula from HREI/MS C20H14N2O2. Furthermore the spectrum exhibited fragments at m/z 285 and 275 (M+ - CO and M+ - CHO) confirming the presence of carbonyl and aldehyde groups respectively. The 1HNMR spectrum of NL/2 (Table 7) displayed two doublets and two triplets in the aromatic region, besides two methylenes indicative of a tetrahydro-β-carboline nucleus (ring A, B and C) in addition to δ-lactam ring (D). The presence of two other aromatic protons at

δH 8.71 (1H, d, J = 7.3 Hz) and δH 7.55 (1H, t, J = 7.3 Hz) could be assigned to H-17 and H-18, respectively, whiles the third proton at δH 8.05 (1H, d, J = 7.3 Hz) could be assigned to H-19.

94

The 13CNMR and DEPT spectra of naucleficine 115 indicated a total of twenty carbon signals; two methylenes, eight methines, eight quaternary carbons and two carbonyl carbons (Table 7). Two downfield carbon signals at δC 161.9 (C- 22) and δC 193.6 (C-21) could be assigned to carbonyl group of a δ-lactam ring and an aldehyde unit, respectively. In the HMBC spectrum, correlation of H-14 (δH 8.17) to C-3 (δC 135.3), H-5 (δH 4.54) to C-3 (δC 135.3) and C-22 (δC 161.9) can be observed, thus supporting the connectivity of ring C with ring D (δ-lactam ring).HMBC correlations of H-14 to C-16 (δC 126.4), H-17 to C-22 (δC 161.9), H-18 to C- 15 (δC 129.5) and C-16 indicating connectivity of the δ-lactam ring D with benzene ring (ring E) through C-15 and C-16. Furthermore, the HMBC spectrum showed correlation of H-19 (δH 8.05) to C-21, H-21 (δH 10.23) to C-20 suggesting that the aldehydic moiety is linked to ring E through C-20 (Figure 9). All spectral data and all 13CNMR signals (Table 7) were in agreement with the published data for naucleficine 115 (Mao et al., 1984; Naito et al., 1991). Naucleficine 115 was isolated before from Nauclea officinalis (Naito et al., 1991), but for the first time from Nauclea latifolia (DNP database, 2011). The antileishmanial activity was not reported for this compound before.

95

Structure of naucleficine 115

N O N H 1H-13C NMBC COSY1H-1H O

H

Figure 9. Selected COSY and HMBC Correlations of naucleficine 115

96

Table 7. 1HNMR and 13CNMR spectral data for compound NL/2 115, (MeOD, 1H: 400 MHz, 13C:75 MHz δ (ppm)) compared with the literature data (MeOD, 1H: 400 MHz,13C:100 MHz) (Mao et al., 1984; Naito et al., 1991) 1H 13C H (multiplicity, J in Hz) C Position Experimental Reference Experimental Reference (CDCl3) (DMSO-d6) (CDCl3) (DMSO-d6) NH-1 8.90 (br s) 11.76 (br s) - - 2 - - 128.0 127.8 3 - - 135.3 135.0 5 4.54 (t, 6.9) 4.42 (t, 6.6) 40.9 40.4 6 3.16 (t, 6.9) 3.12 (t, 6.6) 19.9 19.1 7 - - 115.2 114.0 8 - - 126.1 125.4 9 7.57 (d, 7.8) 7.60 (d, 7.2) 119.6 119.3 10 7.17 (t, 7.8) 7.08 (t, 7.8) 120.7 119.6 11 7.31 (t, 7.8) 7.24 (t, 7.7) 125.0 124.0 12 7.46 (d, 7.8) 7.48 (d, 8.1) 111.8 111.9 13 - - 138.4 138.5 14 8.17 (s) 8.11 (s) 96.1 94.8 15 - - 129.5 129.5 16 - - 126.4 125.5 17 8.71 (d, 7.3) 8.53 (d, 7.5) 134.9 134.2 18 7.55 (t, 7.3) 7.62 (t, 7.2) 125.3 125.9 19 8.05 (d, 7.3) 8.24 (d, 7.5) 141.6 138.4 20 - - 135.8 135.5 21 10.23 (s) 10.43 (s) 193.6 192.6 22 - - 161.9 160.7

97

4.2.3 Characterization of NL/3 as strictosamide 44 NL/3 was obtained as yellowish brown amorphous solid soluble in methanol and exhibited optical activity with [α] -40.0º (c 0.1, MeOH). UV spectrum showed absorption bands at 387, 329, 225 and 203 nm. The maximum at 229 nm is characteristic for indole alkaloids (Au et al., 1973). Stretching of conjugated lactam carbonyl and C-O groups were observed at 1653 and 1050 cm−1 respectively in its IR spectrum. HPLC profile showed a very clear peak in both reverse phase and analytical column indicating the purity of this compound. The purity was confirmed further by EI/MS which gave (M+) at m/z 498. High resolution mass showed the molecular formula + C26H30N2O8 (calc. 521.1894). FAB/MS (+ve) showed (M+H) peak at 499. ESI/MS (+ve) displayed a molecular ion peak (M+H)+ at m/z 499 with logic fragmentations, 337, 267, 171 and 144. 1HNMR spectrum of NL/3 (Table 8) showed in the aromatic region similar pattern to NL/2 (naucleficine), besides the presence of two methylenes which were indicative of the presence of a tetrahydro-β-carboline nucleuses (Ring A, B and C). The 13CNMR and DEPT spectra of NL/3 indicated a total of twenty six carbon signals. The carbonyl carbon of the δ-lactam ring appeared at downfield region at δC 167.1 (C-22). From COSY spectrum a broad doublet of methine proton, H-3, at δH 5.10- showed correlation with a pair of geminal methylene proton signal at δH 2.47- 2.51 and δH 2.07 which were assigned to H-14a and H-14b, respectively. These methylene protons also coupled with the methine proton, H-15, at δH 2.80-2.84. Furthermore, correlation of two dd at δH 5.40 and δH 5.35 (H-18a and H-18b, respectively) with an olefinic methine of H-19 at δH 5.68 can be observed from COSY spectrum. H-19 also showed COSY correlation with the allylic proton H-20 (δH 2.27) which in turn also coupled with the methine

98 proton, H-15. HMBC spectrum showed correlation of the geminal protons, 2H- 18, with an allylic methine carbon of C-20 at δC 44.8. HMBC correlation at H-17 (δH 7.40) to C-15 (δC 25.0), C-16 (δC 109.3), C- 21 (δC 98.1) and C-22, H-20 to C-15 and C-16 as well as H-21 to C-15 and C- 17 (δC 149.2) indicating a 3,4-dihydro-2H-pyran (ring E) was attached to the δ-lactam (ring D) (Figure 10). Besides, the 13CNMR spectrum displayed the signal of a glucose moiety. The anomeric carbon C-1’ and H-1’ signals were observed at δC 100.5 and δH 4.60, respectively. HMBC spectrum showed correlation of the acetal proton H-21 at δH 5.44 (1.7 Hz) with C-1’ suggesting a glucose moiety is attached to C-21 (δC 98.1). Similar NMR spectroscopic values were previously reported for strictosamide 44 (Zhang et al., 2001). The configuration of C-21 was deduced to be β by the coupling constant of 7.9 Hz between anomeric proton H-1’ and H-21 (Zhang et al., 2001; Erdelmeier et al., 1991; Sun et al., 2008). For an α configuration, the coupling constant value will be around 1.2 Hz due to the difference in the dihedral angles between both isomers (Sivasothy et al., 2012). From the analysis of the spectroscopic data obtained (Table 8) and comparison with the literature values, the structure of NL/3 was unequivocally established as strictosamide 44. Strictosamide 44 was isolated before from Nauclea Latifolia, Nauclea orientalis (Zhang et al., 2001) and from Rhazya stricta (Apocynaceae) (DNP database, 2011). The anti-inflammatory, antileishmanial and other antiparastic activities of strictosamide were previously reported (Khalid, 2012).

99

Structure of strictosamide 44

9 6 8 5 10 7 4 11 N O 2 13 22 12 N 3 H1 H H 16 17

20 O 1H-13C NMBC 19 21 H 18 O O HO 1' 2' HO OH OH

Figure 10. Selected HMBC Correlations of strictosamide 44

100

Table 8. 1H and 13CNMR spectral data for compound NL/3 44, (MeOD, 1H: 400 MHz, 13C:75 MHz δ (ppm) compared with the literature data (MeOD, 1H: 400 MHz, 13C:100 MHz) (Zhang et al., 2001)

H (multiplicity, J in Hz) C Experimental Experimental Reference Reference (CD3OD) (CD3OD) (CD3OD) (CD3OD) NH-1 - - - - 2 - - 134.8 134.8 3 5.10 (br d) 5.3 (m) 55.1 55.1 5a 4.98 (dd, 5.6, 12.5) 4.93 (dd, 5.0, 12.0) 44.8 44.7 5b 3.14 (dt, 4.6, 12.5) 3.07 (dt, 4.0, 12.0) 6a 2.95-2.97 (m) 2.93 (m) 22.1 22.1 6b 2.69-2.72 (m) 2.53-2.69 (m) 7 - - 110.3 110.3 8 - - 128.7 128.7 9 7.41 (d, 8.1) 7.37 (d, 8.1) 118.7 118.7 10 7.03 (dt, 8.1, 1.0) 6.99 (dt, 8.1, 1.0) 120.2 120.1 11 7.12 (dt, 8.1, 1.0) 7.07 (dt, 8.1, 1.0) 122.5 122.5 12 7.35 (d, 8.1) 7.32 (d, 8.1) 112.3 112.3 13 - - 137.8 137.8 14a 2.47-2.51 (m) 2.44 (m) 27.4 27.3 2.07 (ddd, 13.9, 2.02 (ddd, 13.8, 14b 13.9, 5.9) 13.8, 6.0) 15 2.80-2.84 (m) 2.79 (m) 25.0 24.9 16 - - 109.3 109.2 17 7.40 (s) 7.36 (s) 149.2 149.4 18a 5.40 (dd, 17.1, 1.7) 5.38 (dd, 16.0, 2.0) 120.5 120.5 18b 5.35 (dd, 10.2, 1.7) 5.33 (dd, 10.0, 2.0) 5.68 (ddd, 17.2, 5.64 (ddd, 17.0, 19 134.4 134.4 10.2, 10.2) 10.0, 10.0) 20 2.69-2.72 (m) 2.63-2.69 (m) 44.8 44.7 21 5.44 (d, 1.7) 5.29 (d, 2.0) 98.1 98.1 22 - - 167.1 167.1 1’ 4.60 (d, 7.9) 4.56 (d, 7.9) 100.5 100.5 2’ 2.96-2.99 (m) 2.96 (m) 74.3 74.3 3.26-3.30 3’ 78.3 78.2 (m, overlapped) 4’ 3.19-3.22 (m) 3.16-3.34 (m) 71.4 71.4 3.26-3.30 5’ 78.0 77.9 (m, overlapped) 6a’ 3.88(dd, 11.9, 2.1) 3.85 (dd, 12.0, 1.9) 62.6 62.6 6b’ 3.65(dd, 11.9, 6.1) 3.62 (dd, 12.0, 5.7)

101

4.3 Antileishmanial and cytotoxicity assays For the purpose of drug discovery, many different assays, using either promastigotes or amastigotes have been developed to screen plant extracts and pure compounds against leishmania. Each of the different assays has advantages and drawbacks. The most straightforward assays use free-living parasites to assess the effect of compounds on cell viability. The main advantage of such assays is that they allow fast and easy screening of bioactive compounds; a feature that is often required for the successful identification of promising antileishmanial hits. Promastigotes have been used routinely for this purpose, but since they represent the insect life-stage, there is the risk of identifying compounds that do not affect the relevant disease-causing life- stage. An alternative is to use axenic amastigotes, i.e., amastigotes that have been adapted to grow outside their host cell in a growth medium that mimics the intracellular conditions. Such amastigotes can be used for straightforward high-throughput screening in a standard growth assay and have the advantage of being more similar to the disease-relevant parasite stage. A number of publications show that screening with promastigotes results in a large set of hits that show no activity against the intracellular parasites (De Rycker et al., 2013). Attempts have been made in the present research to subject the extracts and their fractions to assays targeting both the promastigotes and amastigotes to determine the differences in hits identified by both platforms. Assaying the antileishmanial activity against the promastigotes of Leishmania donovani, Leishmania major and Leishmania tropica of the crude ethanolic extracts of Argemone mexicana and Nauclea latifolia and their respective petroleum ether, chloroform and ethyl acetate fractions as well as the water residue by colorimetric method revealed a remarkable difference in susceptibility of the three leishmania species (Table 9).

102

Table 9. IC50 of the two plants extracts/fractions against the promastigotes of L. donovani, L.major and L.tropica compared with the IC50 of the standard drugs Amphotericin B and Pentamidine by in vitro method (colorimetric)

Extracts/Fractions IC50 (μg/mL± S.D.) IC50(μg/mL± S.D.) IC50(μg/mL±S.D.) L. donovani L. major L. tropica Argemone mexicana Pet. ether fraction 32.57± 0.035 >100 >100 Ethyl acetate fraction 40.45 ± 0.45 51.55 ± 1.91 >100 Chloroform fraction 44.03 ± 0.97 >100 >100 Crude ethanol extract 11.39 ± 0.015 >100 >100 Water residue 23.93 ± 0.34 >100 >100 Nauclea latifolia Pet. ether fraction 48.92 ± 0.08 >100 >100 Ethyl acetate fraction 94.95 ± 0.72 >100 >100 Chloroform fraction. 62.98 ± 0.48 >100 >100 Crude ethanol extract 46.86 ± 0.44 >100 92.52 ± 0.66 Water residue 30.54 ± 1.19 >100 >100 Standard drugs Amphotericin B 2.33 ± 0.20, 00.28 ± 0.05 00.50 ± 0.05 Pentamidine 2.26 ± 0.03 05.09 ± 0.40 08.59 ± 0.05

The crude ethanolic extract of Argemone mexicana exhibited the most prominent activity against the promastigotes of Leishmania donovani (11.39 μg/mL) followed by the water residue (23.93 μg/mL) which is indicative of the hydrophilicity of the bioactive compounds. This most probably can be attributed to quaternary benzophenanthridine alkaloids such as sanguinarine and chelerythrine and protoberberine alkaloids such as berberine which are among the main alkaloids of A. mexicana. Nevertheless, the organic fractions of this taxon exhibited some moderate activity (IC50 ranging between 32.57 -

103

44.03 μg/mL) against the promastigotes of L. donovani indicative of the presence of some other less bioactive secondary metabolites in these fractions whiles the ethyl acetate showed an extended spectrum of activity against L. major (IC50 51.55±1.91 μg/mL) as well as L. donovani (IC50 40.45±0.45 μg/mL).

The crude ethanolic extract and water residue of Nauclea latifolia exhibited weak activity against the promastigote of L donovani with IC50 of 46.86 and 30.54 μg/mL, respectively. All the fractions, however, exhibited far less bioactivity with IC50 ranging between 48.92±0.08 and 94.95±0.72 μg/mL. Interestingly, none of the fractions exhibited any bioactivity against L. major and L. tropica. The presence of this relatively moderate activity of the water residue of N. latifolia might be associated with the presence of the β-carboline alkaloid, strictosamide which is further characterized by its intermediate polarity due to the presence of glucose moiety incorporated into its structure. It worth mentioning at this junction that a similar activity against L. donovani with IC50 of 29.7 μg/mL has already been reported (Khalid, 2012). The standard reference antileishmanial drugs amphotericin B and pentamidine exhibited different activity against the three leishmania species tested. However, both the two standard drugs showed more prominent activity towards L. major and L. tropica with amphotericin B exhibiting potency against Leishmania major and Leishmania tropica when compared with pentamidine in the present study. Apparently most of the activities of the two plants screened were almost confined to the promastigotes stage of L. donovani excluding the ethyl acetate fractions of Argemone mexicana which expressed certain activity towards L. major (IC50 51.55 μg/mL), whiles the ethanolic crude extract of

104

Nauclea latifolia revealed very week bioactivity against L. tropica (IC50 92.52 μg/mL).

The antileishmanial activity of the typical quaternary benzophenanthridine alkaloid such as sanguinarine might be explained in terms of their very strong DNA intercalator (Bai et al., 2006), while the berberine is possessing antimicrobial, antileukemic, anticancer, and topoisomerase inhibitory activities. Berberine is known as a DNA binder and its binding affinities have been extensively characterized (Qin et al., 2006).

The two plant extracts and their respective fractions were also assayed for their activity against the amastigote of L. major and L. tropica but was not test against the amastigote of L. donovani due to the non-availability of L. donovani parasite cells, Table 10 displaying the leishmanicidal activity against the two plant extracts and their fractions against the amastigoate stage of L. major and L. tropica using sodium stibogloconate (Pentostam®) as a standard reference drug.

105

Table 10. IC50 of the extracts/fractions against the amastigotes of L. major and L. tropica compared with the IC50 of the standard drug, sodium stibogloconate (Pentostam®) (13.47 ± 0.17 μg/mL) by macrophages method. Extracts/fractions IC50 (μg/mL± SD.) IC50 (μg/mL± SD.) L. major L. tropica Argemone Mexicana Pet. ether fraction >I00 89.43 ± 1.19 Ethyl acetate fracion 26.32 ± 0.50 >I00 Chloroform fraction 33.95 ± 0.79 >I00 Crude ethanol extract 26.32 ± 0.10 >I00 Water residue //////// 88.75 ± 1.25

Nauclea latifolia Pet. ether fraction 26.97 ± 0.58 91.28 ± 0.65 Ethyl acetate fraction 35.62 ± 0.28 >I00 Chloroform fraction >I00 >I00 Crude ethanol extract 28.69 ± 1.06 88.94 ± 1.06 Water residue 26.76 ± 0.37 >I00

Apparently, Leishmania major was much more susceptible to the most crude extracts and organic fractions of Argemone mexicana and Nauclea latifolia when compared with Leishmania tropica (Table 10). Both the crude extract and the ethyl acetate fractions of A. mexicana exhibited identical specific activity with IC50 of 26.32±0.10 μg/mL towards L. major and almost no activity against L.a tropica. Among the purified isolated alkaloids of N. latifolia, strictosamide inhibited the promastigotes of both L. major and L. tropica with IC50 of 77.67±1.11 and 80.40±3.00 μg/mL), respectively. The structurally related β-carboline alkaloid, naucleficine, however, showed no inhibition as shown in Table 11. Similarly the isoquinoline alkaloid, norchelerythrine, isolated from A. mexicana, exhibited no leishmanicidal activity against L. major and L. tropica (Table 11).

106

Table 11. IC50 of the pure compounds isolated from the two plants against Leishmania major and Leishmania tropica compared with the IC50 of the standard drugs Amphotericin B (0.29±0.05 μg/mL) and Pentamidine (5.09±0.09 μg/mL) by in vitro method (colorimetric) Compounds IC50 ( μg/mL± SD.) IC50 ( μg/mL± SD.) L. major L. tropica Strictosamide 77.67±1.11 80.40 ± 3.00 Naucleficine >100 >100 β-sitosterol >100 >100

Norchelerythrine >100 >100

The relatively bioactive lieshmanicidal alkaloid, strictosamide was subjected to cytotoxicity test against CC-1, MDBK and 3T3 cell lines and exhibited no cytotoxicity with IC50 more than the highest dose used in this assays (20 µg/mL) against the three cell lines.

4.4 Docking of the isolated compounds against leishmania enzymes In the current docking study, four natural compounds were docked against 14 different leishmania enzymes, the table below shows the docking binding energies of the top ranked molecules with the corresponding enzymes. Strictosamide has the relatively strong binding to phosphodiesterase B1 (LmajPDEB1) (PDB: 2R8Q) and Uridine 5'-monophosphate synthase (LdUMPS) (PDB: 3QW4) among all the isolated alkaloids with binding energies of -12.8 and -11.0, respectively. The pentacyclic triterpene, β- sitosterol, however was the only compound among the isolated triterpenes to exhibit selective binding to heat-shock protein (PDB: 3U67) and Tyrosyl- tRNA synthetase (LmajTyrRS) (PDB: 3P0H) with binding energies of -13.1 and -10.9, respectively. The docking results revealed that β-Sitosterol 114 relays basically on the hydrophobic interactions with the greasy residues

107 forming the enzyme binding site, namely; Ala37, Ala40, Met83, Leu92, Val121, Phe123, and Ile171. Besides, a single hydrogen bond between the hydroxyl group attached to position 3 of the compound and Gly120 at a distance of 1.62 Å. Table 12. Docking Binding free energies of the top ranked compounds with leishmania enzymes.

Target PDB Top Ranked Docking Compound Energy Kcal/mol 1. Heat-Shock protein 3u67 β-Sitosterol -13.1 Phosphodiesterase B1 2R8Q Strictosamide -12.8 2. (LmajPDEB1) Uridine 5'-monophosphate 3QW4 Strictosamide -11 3. synthase (LdUMPS) Tyrosyl-tRNA synthetase 3P0H β-Sitosterol -10.9 4. (LmajTyrRS) 5. Aldolase (Lmex ALD) 1SVV β-Sitosterol -9.9 Pteridine reductase (LdPTR, 2XOX β-Sitosterol -8.7 6. LmajPTR1) Glyceraldehyde-3-phosphate 1I33 Naucleficine -8.3 7. dehydrogenase (Lmex GAPDH) Methionyl-tRNA synthetase 3KFL Berberine -8.2 8. (LmajMetRS) Phosphomannomutase 2I54 Strictosamide -8 9. (LmexPMM) Adenine phosphoribosyltransferase 1MZV Strictosamide -7.7 10. (LdAPRT) Triosephosphate isomerase (Lmex 1IF2 Strictosamide -7.7 11. TIM) Deoxyuridine triphosphate nucleotidohydrolase 2YB0 Strictosamide -7.6 12. (LmajdUTPase) Phosphoglycerate mutase (Lmex 3IGY β-Sitosterol -6.2 13. iPGAM) Dihydroorotate dehydrogenase 3C61 Berberine -6 14. (LmajDHODH)

108

A B

Figure 11. Interactions of β-Sitosterol and LmajHSP90. (A) A picture generated by LigX module of MOE software; protein residues are coloured according to their nature. The map shows the 2D visualization of the interactions, represented by the hydrogen bonding (green dots) between the hydroxyl group of the compound and the amino acid Gly120, and van der waal contacts with the hydrophobic residues (coloured green). (B) 3D representation of compound β-Sitosterol within the active site of LmajHSP90 (carbons are in pink, nitrogens in blue and oxygens in red), H-bonds are shown as dark pink dotted line.

A B

Figure 12. Interactions of β-Sitosterol and LmajPDEB1. (A) 2D view, in which the molecule is interacted the enzyme through 3 hydrogen bonding with Val839, Val836 and van der waal contacts with the hydrophobic residues (B) 3D representation of compound β-Sitosterol within the active site of LmajPDEB1)

109

5. CONCLUSIONS AND RECOMMENDATIONS

5.1 CONCLUSIONS

Secondary metabolites that exist in plants are very important natural products that have proven to have many biological activities to enable using them as drugs or lead to drug discovery. Alkaloids are the most useful secondary metabolites as many reports have been published for evaluation of their importance and biological activities as antiplasomodial, antibacterial, anti- inflammatory, antiparasitic and others. There are many reports for antileishmanial activity of many alkaloids from plants. Isolation and structure elucidation of alkaloids or any other class of secondary metabolites are very difficult jobs and need to be meticulous, patient, caring, precise, follow-up and hard worker with the aid of high and modern techniques, as well as using large amount of plant materials for good yield and increase the probability to isolate new or novel components that may have different mechanism of action and lead to good achievements in drug discovery for treatment of various diseases. A. mexicana and N. latifolia literature revealed the presence of many types of alkaloids that have great pharmacological importance. The alkaloids that were isolated in the current study are known, but some of them were isolated for the first time from these plants and/or their families, so they can be considered as new sources.

110

In vitro and in vivo Antileishmanial activity of these alkaloids or extracts containing them with other secondary metabolites have been evaluated in this study against three species of leishmania, L. major and L. tropica, the cusative agents for coutanious leishmaniasis as well as L. donovani which is responsible for the fatal visceral leishmaniasis. Good results have been achieved, some extracts showed significant activity with IC50 close to the IC50 of the standard drugs that were used in the experiments, amphotericin B, pentamidine and sodium stibogloconate in both the in vitro study by colorimetric method and in vivo using macrophage cells.

111

5.2 RECOMMENDATIONS

More attention should be taken for the neglected tropical diseases, as it is fatal and cause death of millions people worldwide annually. There are still huge amount of active components from plants waiting to be discovered since plants showed very effectiveness results in treating various diseases from ancient time. With great attention and focusing, it will be achievable to have effective drugs for the lethal neglected tropical diseases and principally for leishmaniasis. Early diagnosis for leishmaniasis lead to control and cure the disease, such centers should be offered in countries where the leishmanisis are endemic. Alkaloids revealed antiparasitic properties and sure that will lead to effective drugs with more in vivo trials, as not all reported alkaloids were involved in clinical trials. Working in groups gives more valuable achievements in this direction; different information in biology and medicine as well as chemistry will shorten the time and allow going safely for the target, drug discovery. International courses update us about new approach in treating these diseases worldwide, the real risk and size of the problem as eminent scientists made appreciated efforts to treat these diseases. Efforts should be continue and intensify to reach the goal. Further work should be done to evaluate the in vivo antileishmanial activity using more strains and species of leishmania parasites and use these active components in animal trials as there were few published reports in this part and to date there is no effective drug for treatment of leishmaniasis.

112

Spectroscopic techniques, UV, IR, NMR and MS are the heart of structure elucidation, such machines should be accessible for researchers, otherwise students must be sent abroad for research to learn about these techniques and accomplish valuable work.

113

6. References

Abbah, J., Amos, S., Chindo, B., Ngazal, I., Vongtau, H.O., Adzu, B., et al., (2010). Pharmacological evidence favouring the use of Nauclea latifolia in malaria ethnopharmacy: Effects against nociception, inflammation, and pyrexia in rats and mice. Journal of Ethnopharmacology. 127 (1), 85–90.

Ali, H., Konig, G.M. Khalid, S.A., Wright, A.D., Kaminsky, R. (2002) Evaluation of selected Sudanese medicinal plants for their in vitro activity against hemoflagellates, selected bacteria, HIV-1-RT and tyrosine kinase inhibitory, and for cytotoxicity. Journal of Ethnopharmacology. 83, 219-228.

Agyare, C., Mensah, A.Y., Osei-Sante, S., (2006). Antimicrobial activity and phytochemical studies of some medicinal plants from Ghana. Boletin Latinoamericano y del Caribe de plantas medicinales y aromaticas. 5 (6), 113-117.

Amos, S., Abbah, J., Chindo, B., Edmond, I., Binda, L., Adzu, B., et al., (2005). Neuropharmacological effects of the aqueous extract of Nauclea latifolia root bark in rats and mice. Journal of Ethnopharmacology. 97 (1), 53–57.

Araujo, C.A.C., Alegrio, L.V., and Leon, L.L. (1998). Antileishmanial activity of compounds extracted and characterized from Centrolobium sclerophyllum. Phytochemistry. 49 (3), 751–754.

Au, T. Y., Cheung, H. T. and Sternhell, S. (1973). New corynanthe alkaloids from Strychnos angustiflora. J Chem Soc Perkin. 1 (1), 13-16.

Ayeleso, A.O., Oguntibeju, O.O., Brooks, N.L., (2014). In Vitro Study on the Antioxidant Potentials of the Leaves and Fruits of Nauclea latifolia. The Scientific World Journal. 2014, 1–8.

Bai, Li-Ping, Zhao Zhong-Zhen, Cai Zongwei, Jiang Zhi-Hong (2006). DNA-binding affinities and sequence selectivity of quaternary benzophenanthridine alkaloids sanguinarine, chelerythrine, and nitidine, Bioorganic & Medicinal Chemistry. 14 (16), 5439–5445.

Barata, L.E., Santos, L.S., Ferri, P.H., Phillipson, J.D., Paine, A., and Croft, S.L. (2000). Anti-leishmanial activity of neolignans from Virola species and synthetic analogues. Phytochemistry. 55 (6), 589–595.

114

Barral-Netto, M., Von Sohsten, R.L., Teixeira, M., Conrado dos Santos, W.L., Pompeu, M.L., Moreira, R.A., et al., (1996). In vivo protective effect of the lectin from Canavalia brasiliensis on BALB/c mice infected by Leishmania amazonensis. Acta Tropica. 60 (4), 237–250.

Benthe, H.F. (1956). Antifibrillary action of quinidine and similar substances. Naunyn- Schmiedeberg's Archives of Pharmacology. 229, 82-91.

Bentley, K.W. (1998). The isoquinoline alkaloids. CRC Press: London.

Berger, I., Passreiter, C.M., Caceres, A., and Kubelka, W. (2001). Antiprotozoal activity of Neurolaena lobata. Phytotherapy Research. 15 (4), 327–330.

Berger, M., Murugi, J., Buch, E., IJsselmuiden, C., Moran, M., Guzman, J., et al., (2010) Strengthening pharmaceutical innovation in Africa. Council on Health Research for Development (COHRED); New Partnership for Africa’s Development (NEPAD).

Bhattacharjee, I., Chatterjee, S.K., Chandra, G., (2010). Isolation and identification of antibacterial components in seed extracts of Argemone mexicana L. (Papaveraceae). Asian Pacific Journal of Tropical Medicine. 547-551.

Bravo B, J.A., Sauvain, M., Gimenez T.A., Muñoz O.V., Callapa, J., Le Men-Olivier, L., et al., (1999). Bioactive phenolic glycosides from Amburana cearensis. Phytochemistry. 50 (1), 71–74.

Carvalho, P.B., and Ferreira, E.I. (2001) Leishmaniasis phytotherapy. Nature’s leadership against an ancient disease. Fitoterapia. 72.599-618.

Chan-Bacab, M.J., and Peña-Rodríguez, L.M. (2001). Plant natural products with leishmanicidal activity. Natural Product Reports. 18 (6), 674–688.

Chang, Y.C., Chang, F.R., Khalil, A.T., Hsieh, P.W., Wu, Y.C., (2003a). Cytotoxic benzophenanthridine and benzylisoquinoline alkaloids from Argemone mexicana. Zeitschrift fuer Naturforschung, C: Journal of Biosciences. 58, 521-526.

Chang, Y.C., Hsieh, P.W., Chang, F.R., Wu, R.R., Liaw, C.C., Lee, K.H., et al., (2003b). Two new protopines argemexicaines A and B and the anti-HIV alkaloid 6-acetonyl- dihydrochelerythrine from formosan Argemone mexicana. Planta Medica. 69, 148-152.

115

Choudhary, M.I., Yousuf, S., Ahmed, S., Kan, S., Yasmeen and Atta-ur-Rahman (2005). Antileishmanial physalins from Physalis minima, Chemistry and Biodiversity, 2 (9), 1164- 1173.

Choy, C.S., Cheah, K.P., Chiou H.Y., Li, J.S., Liu, Y.H., Yong, S.F., et al., (2008). Induction of hepatotoxicity by sanguinarine is associated with oxidation of protein thiols and disturbance of mitochondrial respiration. Journal of Applied Toxicology. 28, 945-956.

Cordell, G.A. (1981). Introduction to Alkaloids. A Biogenetic Approach. John Wiley & Sons: New York.

Dash, G.K., and Murthy, P.N. (2011). Evaluation of Argemone mexicana Linn. Leaves for wound healing activity. Journal of Natural Product and Plant Resources. 1 (1), 46–56.

Da Silva, S.A.G., Costa, S.S., Mendon cca, S.C.F., Silva, E.M., Moraes, V.L.G., and Rossi- Bergmann, B. (1995). Therapeutic effect of oral Kalanchoe pinnata leaf extract in murine leishmaniasis. Acta Tropica. 60 (3), 201–210.

Delorenzi, J.C., Attias, M., Gattass, C.R., Andrade, M., Rezende, C., Da Cunha Pinto, A., et al., (2001). Antileishmanial Activity of an Indole Alkaloid from Peschiera australis. Antimicrobial Agents and Chemotherapy. 45 (5), 1349–1354.

De Rycker, M., Hallyburton.I., Thomas, J., Campbell, L., Wyllie, S., Joshi, D. Cameron, S., . Gilbert, I.H. . Wyatt, PG., Frearson, JA., Fairlamb, AH. and Gray, DW. (2013) Comparison of a High-Throughput High-Content Intracellular Leishmania donovani Assay with an Axenic Amastigote Assay, Antimicrobial Agents and Chemotherapy. 57 (7), 2913–2922.

Dewick, P. M., (2002) Medicinal Natural Products- A biosynthetic Approach, 2nd edition, John Wiley & Sons, Lt.

Diallo, D., Graz, B., Falquet J., Traoré A.K., Giani S., Mounkoro P.P., et al., (2006). Malaria treatment in remote areas of Mali: use of modern and traditional medicines, patient outcome. Transactions of the Royal Society of Tropical Medicine and Hygiene. 100, 515- 520.

Díaz Chávez, M. L. (2009). Isolation and characterization of (S)-chelianthifoline synthase,(S)-stylopine synthase and two FAD oxidases from a cDNA library from Argemone mexicana. Halle (Saale), Univ., Diss., 2009.

Dictionary of Natural Product database (DNP), (2011).

116

Fournet, A., Barrios, A.A., Muñoz, V., Hocquemiller, R., and Cavé, A. (1993). Effect of some bisbenzylisoquinoline alkaloids on American Leishmania sp. in BALB/c mice. Phytotherapy Research. 7 (4), 281–284.

Eichler, A.W., (1865). Papaveraceae. In: Martius, C.F., editor. Flora Brasiliensis: enumeratio plantarum. J. Cramer: Weinhem; 313-316.

Erdelmeier, C. A. J., Wright, A. D., Orjala, J., Baumgartner, B., Rali, T. & Sticher, O. (1991). New Indole Alkaloid Glycosides from Nauclea orientalis. Planta Med. 57(02), 149-152.

Elford, B.C., (1986). L-Glutamine influx in malaria-infected erythrocytes: a target for antimalarials? Parasitology Today. 2, 309-312.

Elegami, A.A., Al Magboul, A.Z., Omer, M.E., El Tohami, M.S., (2001). Sudanese plants used in folkloric medicine: screening for antibacterial activity. Fitoterapia. 72 (7), 810-817.

El Ghazali, G.B., El Tohami, M.S., El Egami, A.B., (1994). Medicinal Plants of the Sudan. Part III. Medicinal Plants of the White Nile Province. Khartoum University Press, Sudan.

El Ghazali, G.B., El Tohami, M.S., El Egami, A.B., Abdalla, W.S., Mohammed, M.G., (1997). Medicinal Plants of the Sudan. Part IV. Medicinal Plants of Northern Kordofan. Omdurman Islamic University Press, Khartoum, Sudan.

El Kamali, H.M., El Khalifa, K.F., (1997). Treatment of malaria through herbal drugs in the Central Sudan. Fitoterapia. 68 (6), 527-528.

El Kamali, H.M., El Khalifa, K.F., (1999). Folk medicinal plants of riverside forests of the southern Blue Nile district, Sudan. Fitoterapia. 70, 493-497.

El Kamali, H.M., Khalid, S.A., (1996). The most common herbal remedies in Central Sudan. Fitoterapia. 57, 301-306.

El Tahir, A., Ibrahim, A.M, Satti, G.M.H., Theander, T.G., Kharazmi, A., Khalid, S.A., (1998). The potential antileishmanial activity of some Sudanese medicinal plants. Phytotherapy Research. 12 (8), 576-579.

El Tahir, A., Satti, G.M., Khalid, S.A. (1999a). Antiplasmodial activity of selected Sudanese plants with emphasis on Acacia nilotica. Phytotherapy Research. 13, 474-478.

117

El Tahir, A., Satti, G.M., Khalid, S.A., (1999b). Antiplasmodial activity of Sudanese plants with special emphasis on Maytenus senegalensis. Journal of Ethnopharmacology. 64, 227- 233.

Graz, B., Willcox, M.L., Diakite, C., Falquet, J., Dackuo, F., Sidibe, O., et al., (2009). Argemone mexicana decoction versus artesunate-amodiaquine for the management of malaria in Mali. Transactions of the Royal Society of Tropical Medicine and Hygiene. 104 (1), 33–41.

Habtemariam, S., (2003). In vitro antileishmanial effects of antibacterial diterpenes from two Ethiopian Premna species: P. schimperi and P. Oligotricha, Bio Med Central Pharmacology. 3, 1-6.

Hakim, S.A. (1954). Argemone oil, sanguinarine, and epidemic-dropsy glaucoma. The British Journal of Ophthalmology. 38 (4), 193.

Hazra, B., Ghosh, R., Banerjee, A., Kirby, G.C., Warhurst, D.C., Phillipson, J.D., (1995), In vitro antiplasmodial effects of diospyrin, a plant-derived naphthoquinoid, and a novel series of derivatives. Phytotherapy Research. 9 (1),72–74.

Heywood, V.H. (1993). Flowering plants of the world. Oxford University Press: Oxford.

Hussein, G., Miyashiro, H., Nakamura, N., Hattori, M., Kawahata, T., Otake, T., et al., (1999). Inhibitory effects of Sudanese plant extracts on HIV-1 replication and HIV-1 protease. Phytotherapy Research. 13 (1), 31-36.

Iranshahi, M., Arfa, P., Ramezani, M., Jaafari, M.R., Sadeghian, H., Bassarello, C., Piacente, S., et al., (2007). Sesquiterpene coumarins from Ferula szowitsiana and in vitro antileishmanial activity of 7-prenyloxycoumarins against promastigotes. Phytochemistry. 68 (4), 554–561.

Israilov, A., Yunusov, M.S. (1986). Alkaloids of four species of Argemone. Chemistry of Natural Compounds. 22, 189-192.

Iwasa, K., Kim, H.S., Wataya Y., Lee, D.U. (1998). Antimalarial activity and structure- activity relationships of protoberberine alkaloids. European Journal of Medicinal Chemistry. 33, 65-69.

Iwasa, K., Nishiyama, Y., Ichimaru, M., Moriyasu, M., Kim, H.S., Wataya, Y., et al., (1999). Structure-activity relationships of quaternary protoberberine alkaloids having an antimalarial activity. European Journal of Medicinal Chemistry. 34, 1077-1083.

118

Jyothirmayi, N., and Prasad, S.H.K.R., (2012) Efficacy of methanol and chloroform extracts of Euphorbia hirta Linn. in inhibiting pathogens. Journal of Chemical, Biological and Physical Sciences. 2 (1), 334-337. Kan, S., Chen, G., Han, C., Chen, Z., Song, X., Ren, M., et al., (2011). Chemical constituents from the roots of Xanthium sibiricum. Natural Product Research. 25 (13), 1243–1249.

Karou, S.D., Tchacondo, T., Ilboudo, D.P., Simpore, J. (2011). Sub Saharan Rubiaceae: A review of their traditional uses, phytochemistry and bioligical activities. Pakistan journal of biological sciences. 14 (3), 149-169.

Kayser, O., Kiderlen, A.F., and Croft, S.L. (2002). Natural products as potential antiparasitic drugs. Studies in Natural Products Chemistry. 26, 779–848.

Khalid, H., Abdella, W., Abdelgadir, H., Opatz, T., Efferth, T., (2012): Gems from traditional north-African medicine: medicinal and aromatic plants from Sudan. Natural Product Bioprospect. 2, 92–103.

Khalid, S.A., Farouk, A., Geary, T.G., Jensen, J.B., (1986). potential antimalarial candidates from African plants: an in vitro approach using Plasmodium falciparum. Journal of Ethnopharmacology. 15 (2), 201-209.

Khalid, S.A., Duddeck, H., Gonzalez Sierra, M., (1989). Isolation and characterization of the antimalarial agent of the Neem tree, Azadirachta indica. Journal of Natural Products. 52, 922-927.

Khalid, S.A., Friedrichsen, G.M., Kharazmi, A., Theander, T., Christensen, S.B., Olsen, C.E., (1998). Limonoids from Khaya senegalensis, characterization of 2,6-dihydroxy- fissinolide as a new limonoid. Phytochemistry. 49, 1796-1772.

Khalid, S.A., (2012). “Natural products-based drug discovery against neglected diseases with special reference to African natural resources''. In: Chibale, K., Masimirembwa, C., and Davies-Coleman, M., (eds.), Drug Discovery in Africa - Impacts of Genomics, Natural Products, Traditional Medicines, Insights into Medicinal Chemistry, and Technology Platforms in Pursuit of New Drugs, 211-237. New York, Berlin, Heidelberg, Springer- verlag.

119

Kolak, U., Topçu, G., Seher Birteksöz, Ötük, G. & Ulubelen, A., (2005). Terpenoids and steroids from the roots of Salvia blepharochlaena. Turkish Journal of Chemistry. 29, 177- 186.

Kosalge, S.B., and Fursule, R.A., (2009). Investigation of ethnomedicinal claims of some plants used by tribals of Satpuda Hills in India. Journal of Ethnopharmacology. 121, 456- 461.

Lewis, K., Ausubel, F.M., (2006). Prospects for plant-derived antibacterials. Nature Biotechnology. 24, 1504-1507.

Leyva-Peralta, M.A., Robles-Zepeda, R.E., Garibay-Escobar, A., Ruiz-Bustos, E., Alvarez- Berber, L.P., Galvez-Ruiz, J.C., (2015). In vitro anti-proliferative activity of Argemone gracilenta and identification of some active components. BMC Complementary and Alternative Medicine. 15 (13), 1-7.

Li, B., Wu, Q., Shi, J., Sun, A., Huang, X., (2005). Effects of protopine on intracellular calcium and the PKC activity of rat aorta smooth muscle. Shengli Xuebao. 57, 240-246.

Li. Y., Wang, S., Liu, Y., Li, Z., Yang, X., Wang, H., et al., (2008). Effect of α- allocryptopine on transient outward potassium current in rabbit ventricular myocytes. Cardiology. 111, 229-236.

Maikai, V.A., and Kobo, P.I. (2008). Preliminary studies on the in vitro antitrypanosomal activity of aqueous and methanolic crude extracts of stem bark of Nauclea latifolia on Trypanosoma congolense. Journal of Medicinal Plants Research. 2 (6), 115–118.

Martin, M.T., Rasoanaivo, L.H., Raharisololalao, A. (2005). Phenanthridine alkaloids from Zanthoxylum madagascariense. Fitoterapia. 76 (6), 590–593.

Mao, L., Xin, L., Dequan, Y., (1984) Alkaloids of Nauclea officinalis. Planta medica. 459- 461. MBG. 2009. Missouri Botanical Garden Tropicos Database. Accessed on May 19, 2009 (http://www.tropicos.org).

Mishra, B.B., Kale, R.R., Prasad, V., Tiwari, V.K., Singh, R.K., (2011) 4. Scope of natural products in fighting against leishmaniasis. Opportunity, Challenge and Scope of Natural Products in Medicinal Chemistry.121-154.

120

Mohana, D.C., Satish, S., Raveesha, K.A. (2008). Antibacterial evaluation of some plant extracts against some human pathogenic bacteria. Advances in Biological Research. 2 (3- 4), 49–55.

Monzote, L., (2009). Current treatment of leishmaniasis: A review. The Open Antimicrobial Agents Journal. 1, 9-19.

Naito, T., Kuroda, E., Miyata, O., Ninomiya, I., (1991). "Photocyclization of Enamides. XXXIV. a practical total synthesis of aromatic yohimboid alkaloids, oxogambirtannine and nauclificine." Chem. Pharm. Bull. 39 (9), 2216-2218.

Ngnokam, D., Ayafor, J.F., Connolly, J.D., Nuzillard, J.M., (2003). Nauclefolinine: a new alkaloid from the roots of Nauclea latifolia. Bulletin of the Chemical Society of Ethiopia. 17 (2), 173-176.

Nworgu, Z.A.M., Onwukaeme, D.N., Afolayan, A.J., Ameachina, F.C., and Ayinde, B.A., (2008). Preliminary studies of blood pressure lowering effect of Nauclea latifolia in rats. African Journal of Pharmacy and Pharmacology. 2 (2), 037–041.

Okwu, D.E., and Uchenna, N.F., (2009). Exotic multifaceted medicinal plants of drugs and pharmaceutical industries. African Journal of Biotechnology. 8 (25), 7271-7282.

Onyeyili, P.A., Nwosu, C.O., Amin, J.D., Jibike, J.I., (2001). Anthelmintic activity of crude aqueous extract of Nauclea latifolia stem bark against ovine nematodes. Fitoterapia. 72, 12- 21.

Osorio, E.J., Robeldo, S.M., Bastida, J., (2008). Chapter 2 Alkaloids with antiprotozoal activity. In The Alkaloids: Chemistry and Biology. Academic Press: London, Elsevier. 66, 113-179.

Perumal, P., Sekar, V., Rajesh, V., Gandhimathi, S., Sampathkumar, R., Shuja Nazimudin K.H., (2010). Invitro Antioxidant Activity of Argemone mexicana Roots. International Journal of PharmTech Research. 2 (2), 1477-1482.

Piacente, S., Capasso, A., De Tommasi, N., Jativa, C., Pizza, C., Sorrentino, L., (1997). Different Effects of Some Isoquinoline Alkaloids from Argemone mexicana on Electrically Induced Contractions of Isolated Guinea-pig Ileum. Phytotherapy Research. 11, 155–157.

121

Poonam, K., and Singh, G.S., (2009). Ethnobotanical study of medicinal plants used by the Taungya community in Terai Arc Landscape, India. Journal of Ethnopharmacology. 123, 167-176.

Qin, Yong , Pang Ji-Yan , Chen Wen-Hua , Cai Zongwei , Jiang Zhi-Hong (2006) Synthesis, DNA-binding affinities, and binding mode of berberine dimers, Bioorganic & Medicinal Chemistry, 14 (1), 25–32.

Rao, G.V., Annamalai, T., Mukhopadhyay. T., Machavolu, S., Lakshmi Madhavi, (2011) "Chemical constituents and melanin promotion activity of Cissus quadranglaris Linn". Research Journal of Chemical Sciences. 1 (2), 25-29.

Reddy, B.U., (2010). Enumeration of Antibacterial Activity of Few Medicinal Plants by Bioassay Method. Journal of Chemistry. 7 (4), 1449–1453.

Rocha, L.G., Almeida, J.R.G.S., Macêdo, R.O., Barbosa-Filho, J.M., (2005). A review of natural products with antileishmanial activity. Phytomedicine. 12 (6-7), 514–535.

Rodriguez-Menchaca, A., Ferrer-Villada, T., Lara, J., Fernandez, D., Navarro-Polanco, R. A., Sanchez-Chapula, J. A., (2006). Block of hERG channels by berberine: mechanisms of voltage and state-dependence probed with site-directed mutant channels. Journal of Cardiovascular Pharmacology. 47, 21-29.

Ross, R., (1903). Note on the bodies recently described by Leishman and Donovan. British Medical Journal. 2 (2237), 1261.

Saha, P., Bhattacharjee, S., Sarkar, A., Manna, A., Majumder, S., Chatterjee, M., (2011). Berberine chloride mediates its Anti-leishmanial activity via differential regulation of the mitogen activated protein kinase pathway in macrophages. PLoS ONE. 6 (4), e18467.

Sakthivadivel, M., and Daniel, T., (2008). Evaluation of certain insecticidal plants for the control of vector mosquitoes viz. Culex quinquefasciatus, Anopheles stephensi and Aedes aegypti. Applied Entomology and Zoology. 43 (1), 57–63.

Schmidt, T.J., Khalid, S.A., Romanha, A.J., Alves, T.M.A., Biavatti, M.W., Brun, R., et al., (2012a). The Potential of Secondary Metabolites from Plants as Drugs or Leads Against Protozoan Neglected Diseases - Part I. Current Medicinal Chemistry. 19 (14), 2128–2175.

Schmidt, T.J., Khalid, S.A., Romanha, A.J., Alves, T.M.A., Biavatti, M.W., Brun, R., et al., (2012b). The Potential of Secondary Metabolites from Plants as Drugs or Leads Against Protozoan Neglected Diseases - Part II. Current Medicinal Chemistry. 19 (14), 2176–2228.

122

Scott, T., (1996). Concise Encyclopedia. Biology. Walter de Gruyter: New York.

Scudiero, D.A., Shoemaker, R.H., Paull, K.D., Monks, A., Tierney, S., Nofziger et al., (1988). Evaluation of a soluble tetrazolium/formazan assay for cell growth and drug sensitivity in culture using human and other tumor cell lines. Cancer Research. 48, 4827– 4833.

Shigemori, H., Kagata, T., Ishiyama, H., Morah, F., Ohsaki, A., Kobayashi, J., (2003). Naucleamides A-E, new monoterpene indole alkaloids from Nauclea latifolia. Chemical and Pharmaceutical Bulletin. 51 (1), 58–61.

Singh, S., Singh, A., Jaiswal, J., Singh, T.D., Singh, V.P., Pandey, V.B., et al., (2010a). Antifungal activity of the mixture of quaternary alkaloids isolated from Argemone mexicana against some phytopathogenic fungi. Archives of Phytopathology and Plant Protection. 43 (8), 769–774.

Singh, S., Singh, T.D., Singh, V.P., Pandey, V.B., (2010b). A new benzylisoquinoline alkaloid from Argemone mexicana. Natural Product Research. 24 (1), 63–67.

Singh, S.K., Pandey, V.D., Singh, A., Singh, C., (2009). Antibacterial activity of seed extracts of Argemone mexicana L. on some pathogenic bacterial strains. African Journal of Biotechnology. 8 (24),7077-7081.

Sivasothy, Y., Hadi, A. H. A., Mohamad, K., Leong, K. H., Ibrahim, H.,Sulaiman, S. F., et al., (2012). Spectaflavoside A, a new potent iron chelating dimeric flavonol glycoside from the rhizomes of Zingiber spectabile Griff. Bioorganic & Medicinal Chemistry Letters. 22(11), 3831-3836.

Sun, J., Lou, H., Dai, S., Xu, H., Zhao, F. & Liu, K. (2008). Indole alkoloidsfrom Nauclea officinalis with weak antimalarial activity. Phytochemistry. 69(6), 1405- 1410.

Taïwe, G.S., Bum, E.N., Talla, E., Dimo, T., Dawe, A., Sinniger, V., et al., (2014). Nauclea latifolia Smith. (Rubiaceae) exerts antinociceptive effects in neuropathic pain induced by chronic constriction injury of the sciatic nerve. Journal of Ethnopharmacology. 151 (1), 445–451.

Vennerstrom, J.L., Lovelace, J.K., Waits, V.B., Hanson, W.L., Klayman, D.L., (1990). Berberine derivatives as antileishmanial drugs. Antimicrobial Agents and Chemotherapy. 34 (5), 918–921.

123

Verma, S.K., Dev, G., Tyagi, A. K., Goomber S., Jain, G.V., (2001). Argemone mexicana poisoning: autopsy findings of two cases. Forensic Science International. 115, 135-141.

Wang, Y.X., Zheng, Y.M., Zhou, X.B., (1996). Inhibitory effects of berberine on ATP- sensitive K+ channel in cardiac myocytes. European Journal of Pharmacology. 316, 307- 315.

Wang, Y.X., Zheng, Y.M., (1997). Ionic mechanism responsible for prolongation of cardiac action-potential duration by berberine. Journal of Cardiovascular Pharmacology. 30, 214- 222.

Willcox, M.L., Graz, B., Falquet, J., Sidibe, O., Forster, M., Diallo, D., (2007). Argemone mexicana decoction for the treatment of uncomplicated falciparum malaria. Transactions of the Royal Society of Tropical Medicine and Hygiene. 101, 1190-1198.

Wright, C.W., Marshall, S.J., Russell, P.F., Anderson, M.M., Phillipson, J.D., Kirby, G.C., et al., (2000). In vitro antiplasmodial, antiamoebic, and cytotoxic activities of some monomeric isoquinoline alkaloids. Journal of Natural Products. 63, 1638-1640.

Yu, L., Sun, A., Wu, Q., Huang, X., (1999). Effects of protopine on physiological characteristics of isolated guinea pig atrium. Zhongguo Yaolixue Tongbao 15, 432-434.

Zhang, Z., ElSohly, H.N., Jacob, M.R., Pasco, D.S., Walker, L.A., Clark, A.M., (2001). New Indole Alkaloids from the Bark of Nauclea orientalis. Journal of Natural Products. 64 (8), 1001–1005. http://www.thelancet.com/series/neglected-tropical-diseases.

124