A combinatorial approach to the search for anticonvulsant agents from gou teng and tian ma

195 529 0
A combinatorial approach to the search for anticonvulsant agents from gou teng and tian ma

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

A COMBINATORIAL APPROACH TO THE SEARCH FOR ANTICONVULSANT AGENTS FROM GOU TENG AND TIAN MA YANG HONG (B. Sc. (Pharm.)), Sichuan Univ. (M. Sc. (Pharm.)), Sichuan Univ. A THESIS SUBMITTED FOR THE DEGREE OF DOCTOR OF PHILOSOPHY DEPARTMENT OF PHARMACY NATIONAL UNIVERSITY OF SINGAPORE 2009 ACKNOWLEDGEMENTS I would like to give my sincere gratitude to my dear supervisor, Assoc. Prof. Chui Wai Keung. What I have learned besides his gentleness, kindness and generosity is the attitude towards scientific research, meticulous, skeptical and always openminded. I think I am lucky enough to be his student, who has been granted such a great freedom to experiment with wild ideas, guided not to lose the direction while exploring, and encouraged to confidently convey research findings in oral presentation. I hope I would be growing up as an independent researcher, since that will be the best gift a student could think of to give to her teacher. I would also like to thank Dr. Low Chian Ming, who helped me to plan my electrophysiological study and initiated so many insightful discussions regarding the results, Dr. Chew Eng Hui, who gave me valuable advice and shared with me her experiences in culturing cell lines, and Mr. Wee Xi Kai, who taught me how to use autodock program. I am grateful to the Pharmacy department lab technicians namely, Ms Ng Sek Eng, Ms Dyah Nanik Irawati and Ms Lye Pey Pey, for all your hard work in providing technical support. More importantly, I wish to express my deep gratitude to National University of Singapore for offering the research scholarship that supports my candidature. In addition, I would like to acknowledge the academic research grant (R148-000-049-112) for providing financial resources for the work carried out. My dear friends, Dr. Ma Xiang, Ms Pauline Ong, Ms Michelle Kok, Dr. Huang Meng, Ms Wang Chunxia, Mr. Zhang Yaochun, Ms Jin Jing, Ms Yang Shili, Mr. Wang Zhe and Ms Sim Hong May, I owe you all a big thank you. You all know how I can easily cry but you may not know I only cried in front of friends. The time spent with you was so precious and would be preserved in my memories. My dear parents and sister, I am indebted to all of you for being my first teachers and my best companions ever. My dearest husband, Mr. Lu Xiaohua, you have fulfilled my life outside the lab, and I am thanking you for always being there for me, appreciating me as I am in spite of my ill-temper and willfulness. This thesis is written for you. i CONTENTS ACKNOWLEDGEMENTS PAGE i CONTENTS ii SUMMARY v ABBREVIATIONS vii LIST OF SCHEMES xi LIST OF FIGURES xii LIST OF TABLES xv 1. INTRODUCTION 1.1. General introduction 1.2. Drug discovery from natural products 1.2.1. Crude, pre-fractionated and pure natural products library 1.2.2. The reductionist and holistic approach in drug discovery from natural products 10 1.2.2.1. Interplay between constituent in natural products 10 1.2.2.2. Bioassay-guided fractionation and systems biology based metabolomic approach 13 1.2.3. A proposition-purifying one single chemical class from natural products 16 1.2.4. Hyphenated instrumentation applied in the analysis of natural products 18 1.3. Two traditional Chinese Medicines – Gou Teng & Tian Ma 21 1.3.1. Phytochemical and pharmacological studies of Gou Teng 21 1.3.2. Phytochemical and pharmacological studies Tian Ma 26 1.4. Antiepilepsy, antiepileptogenesis and neuroprotection 28 1.4.1. Definition for antiepilepsy, antiepileptogenesis and neuroprotection29 1.4.2. In vitro & in vivo models for epilepsy, epileptogenesis and neuroprotection 30 1.5. The role of NMDA antagonism in antiepilepsy, antiepileptogenesis and neuroprotection 33 1.5.1. NMDA antagonism 33 1.5.2. Antagonists at the glycine site of NMDA receptor 34 1.6. Summary 39 1.7. Hypothesis and objectives 40 2. RESULTS & DISCUSSIONS 43 2.1. The standardized extracts derived from Gou Teng and Tian Ma 43 2.1.1. Methodology 43 2.1.2. The standardized alkaloidal extract from Gou Teng 44 2.1.2.1. Preparation of the alkaloidal extract 45 ii 2.1.2.2. Qualitative analysis of the alkaloidal extract by LC-ESI-MS 46 2.1.2.3. Isolation and characterization of single alkaloids from the alkaloidal extract 48 2.1.2.4. Quantitative analysis of the alkaloidal extract by LC-UV 56 2.1.2.5. Conclusions 60 2.1.3. The standardized phenolic extract derived from Tian Ma 62 2.1.3.1. The preliminary purification and analysis on GC-EI-MS 63 2.1.3.2. Isolation and characterization of single phenolic compounds in the EFME 70 2.1.3.3. Synthesis of some phenolic compounds present in Tian Ma 73 2.1.3.3.1. Retro-synthesis of target compounds 74 2.1.3.3.2. Synthesis of HPMP 75 2.1.3.3.3. Synthesis of BHE, HFC and CSD 79 2.1.3.3.4. Synthesis of HMP 81 2.1.3.4. Further purification on MAX cartridge and analysis on LC-UV 82 2.1.3.5. Conclusions 84 2.1.4. Summary 85 2.2. Antiepileptic activity investigation 85 2.2.1. MES and rotorod test 85 2.2.2. Implications for the results of the phytochemical and animal study 91 2.2.2.1. Justifications for choosing the strategy of the purification of the alkaloids and phenols over bioassay-guided fractionation 91 2.2.2.2. Explanations for the lack of antiepileptic activity possessed by the alkaloidal and phenolic extracts 93 2.3. In vitro neuroprotective activity assessed on neuro-cell line 95 2.3.1. Effects on cell viability in the absence of neurotoxins 96 2.3.2. Effects on cell viability in the presence of hydrogen peroxide 98 2.3.3. Effects on cell viability in the serum-deprived medium 99 2.4. Design and synthesis of potential antagonists at Gly/NMDA receptor and in vivo neuroprotective activity 103 2.4.1. Design of novel antagonists of Gly/NMDAR 103 2.4.2. Synthesis of 2-oxindole derivatives 107 2.4.2.1. Synthesis of 3-[(substituted furan-2-yl)methylidenyl] 2-oxindoles 107 2.4.2.2. Synthesis of substituted-3-benzoyl (furoyl)-2-oxindoles 110 2.4.3. Neuroprotective activity evaluation on SH-SY5Yneuroblastoma cell line 115 2.4.3.1. Effects of 2-oxindole derivatives on cell viability in the absence of neurotoxins 115 2.4.3.2. Effects of 2-oxindole derivatives on cell viability in the presence of hydrogen peroxide 116 2.4.3.3. DPPH free radical scavenging test 118 2.4.4. Electrophysiological characterization of 3112 on Xenopus oocytes expressing NR1/NR2B 120 2.4.5. Conclusions 125 3. CONCLUDING REMARKS 127 4. EXPERIMENTAL 134 4.1. General 134 4.2. Phytochemistry 135 iii 4.2.1. Prepare the standardized extracts 135 4.2.1.1. The alkaloidal extract from Gou Teng 135 4.2.1.2. The phenolic extract from Tian Ma 135 4.2.2. Instrumental analysis of the extracts 136 4.2.2.1. Qualitative analysis of the alkaloidal extract by LC-ESI-MS 136 4.2.2.2. Quantitative analysis of the alkaloidal extract by LC-DAD 137 4.2.2.3. Qualitative analysis of the EFME by GC-EI-MS 137 4.2.2.4. Quantitative analysis of the EFME by LC-DAD 138 4.2.3. Spectroscopic analysis of the isolated compounds 139 4.3. Computational method – Molecular docking 142 4.4. Synthetic chemistry 143 4.4.1. Synthetic procedures 143 4.4.2. Characterization data of synthesized compounds 149 4.4.3. X-ray crystallographic analysis of compound 18 162 4.5. Bioactivity investigations 163 4.5.1. DPPH free radical scavenging test 163 4.5.2. Determination of total free phenolic compounds by Folin-Ciocalteu test 164 4.5.3. MES and Rotorod test 164 4.5.4. Cell culture and cell viability test 165 4.5.5. Electrophysiological study of compound 20 (3112) on Xenopus oocytes expressing NR1/NR2B 166 4.5.6. Statistics 167 5. BIBILIOGRAPHY 168 iv SUMMARY Although current antiepileptic drugs can effectively relieve most of the epileptic patients from episodes of seizure, still 40% of the patients are refractory to the current therapeutics. Hence development of new antiepileptic agents is urgently needed. Gou Teng and Tian Ma are two well-known Traditional Chinese Medicines (TCMs), widely used in oriental medicine to treat epilepsy, hypertension and migraine, but their scientific evidences to support their therapeutic applications in the abovementioned areas are limited. NMDA (N-methyl-D-aspartic acid) antagonism is an important mechanism for many successful antiepileptic and neuroprotective agents, such as MK-801, CPP, ketamine etc. Nowadays NMDA receptor antagonism still remains as an effective strategy for discovery of new antiepileptic and neuroprotective agents. Our aim is to search for active small molecules or molecular combinations from Gou Teng and Tian Ma, and to design and synthesize new NMDA receptor antagonists for the benefit of controlling seizures and protecting against neuronal damages. Conventional extraction and purification techniques were employed to obtain bioactive fractions, which were subsequently subjected to careful qualitative and quantitative chemical analysis. The standardized fractions, derived from Gou Teng and Tian Ma respectively, were tested on the maximal electroshock-induced seizure (MES) model in mice and on cell cultures that were inflicted by either hydrogen peroxide or serum-deprivation. The indole-2-one (2-oxindole) template was utilized to prepare a series of 3-substituted derivatives which were later tested on cell cultures inflicted by hydrogen peroxide and on voltage-clamped Xenopus Oocytes expressed NR1/NR2B NMDA receptor. v The acquired fractions either alone or in combination did not prevent MES induced seizure activities on mice, but the phenolic fraction derived from Tian Ma was found to be rescuing cells from serum-withdrawal induced apoptosis. Two libraries based on 2-oxindole derivatives were synthesized with acceptable yields, and one of the libraries was found to be acting as free radical scavengers to protect against cell death inflicted by hydrogen peroxide. One synthesized compound, namely compound 20 (3112), was found to be a weak agonist acting at the glycine site of NMDA receptor based on the electrophysiological study on Xenopus Oocytes. Therefore, in this project it was found that Gou Teng and Tian Ma might be used more as neuroprotectants rather than as antiepileptic agents. In addition, some of the synthesized 2-oxindole derivatives that were initially designed as antagonists of Gly/NMDAR were acting as neuroprotectants against oxidative stress. In this project, the endeavour to search for potential antiepileptic and neuroprotective agents was made on both natural products and synthetic compounds. NMDA antagonism which was one of the mechanisms for Gou Teng to protect neuronal damage and also the rational basis on which the 2-oxindoles were synthesized served as the link. This was a trial that combined the passive process of isolating constituents from natural products and the active participation of design and synthesis of new compounds based on a well-defined target; this was an experiment that combined the diversified structures found in natural products and the synthetic library with limited structure diversity. It was believed that the most fruitful discovery of drugs should rely on both natural products and organic synthesis. Key words: Antiepileptic agents; Gou Teng (Ramulus Uncariae cum uncis); Tian Ma (Rhizoma Gastrodiae); NMDA receptor; 2-oxindole derivatives. vi ABBREVIATIONS 2CI 2-Carboxyindoles 4HA 4-Hydroxybenzyl Alcohol 4HD 4-Hydroxybenzaldehyde 5-HT 5-Hydroxtryptamine AEDs Antiepileptic Drugs AKU Akuammigine BBB Blood Brain Barrier BHE Bis-(4-hydroxybenzyl) Ether CE Collision Energy CI Combination Index CNS Central Nervous System CNMR Carbon Nuclear Magnetic Resonance COR Corynoxeine CSD Cirsiumaldehyde CT Corynantheine CTHQ 2-Carboxytetrahydroquinolines CUR Curtain Gas DAD Diode Array Detector DCKA 5,7-Dichlorokynurenic Acid DE Diethyl Ether DEPT Distortionless Enhanced by Polarization Transfer DHPM 4,4’-Dihydroxyphenyl Methane DP Declustering Potential DPPH 2,2-Di(4-Tert-Octylphenyl)-1-Picrylhydrazyl vii EAA Excitatory Amino Acid EC50 Half-Effective Concentration) EDRF Endothelium-Derived Relaxing Factor EFME Ether Fraction of Methanolic Extract EGCG (-)-Epigallocatechin Gallate EI Electron Impact EIMS Electron Ionization MS ELSD Evaporative Light Scattering Detector EMS Enhance Q3 Single MS EP Entrance Potential ESI Electrospray Ionization FHPE 1-Furan-2-Yl-2-(4-Hydroxy-Phenyl)-Ethanone GABA γ-Aminobutyric Acid GC Gas Chromatography Gly/NMDAR Strychnine-Insensitive Glycine Site of NMDA Receptor GME Geissoschizine Methyl Ether GS1 Ion Source Gas HBAD 3-Hydroxy-1H-1-Benzazepine-2,5-Diones HFC 5-(4-Hydroxy-Benzyloxymethyl)-Furan-2-Carbaldehyde HME 4-Hydroxybenzyl Methyl Ether HMFC 5-Hydroxymethyl-Furan-2-Carbaldehyde HMP 2,4-Bis[(4-Hydroxyphenyl)Methyl]-Phenol HMQC Heteronuclear Multiple Quantum Coherence HNMR Proton Nuclear Magnetic Resonance HPLC High Pressure Liquid Chromatography viii HPMP 4-((4-hydroxymethyl)phenoxy)methyl) phenol HQ 4-Hydroxy-2-Quinolones HRMS High Resolution MS HS Hirsutine HT Hirsuteine HTS High-Throughput Screening IR Infrared Spectroscopy ISO-RHY Isorhynchophylline JNK C-Jun N-Terminal Kinases KA Kynurenic Acid KI Dissociation Constant LC Liquid Chromatography LEV Levetiracetam MCAO Middle Cerebral Arterial Occlusion MDRs Multidrug Resistance Pumps MES Maximal Electroshock-Induced Seizure MPP+ 1-methyl-4-phenylpyridinium cation MS Mass Spectrometry MTT 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide MVDA Non-linear Multivariate Data Analysis MW Molecular Weights NCEs New Chemical Entities NGF Neuronal Growth Factor NMDA N-methyl-D-aspartic acid NMR Nuclear Magnetic Resonance ix Yellow crystals, yield 95%, mp: 141-143℃; 1H NMR (300MHz, CDCl3) δ (in ppm) 14.2 (s, 1H, OH), 8.32-8.37 (dd, 1H, H-4, J = 0.75 & 8.3 Hz), 8.23-8.26 (dd, 1H, H-7, J = 1.1 & 7.9 Hz), 7.82 (d, 1H, H-4’, J = 1.5 Hz), 7.38-7.39 (d, 1H, H-2’, J = 3.4 Hz), 7.18-7.30 (m, 2H, H-5 & 6), 6.69-6.71 (dd, 1H, H-3’, J = 1.5 & 3.4 Hz), 2.78 (s, 3H, COCH3). 13 C NMR (75MHz, CDCl3) δ (in ppm) 174.0, 170.5, 159.7, 148.3, 146.2, 135.3, 126.6, 124.7, 121.7, 121.3, 118.3, 116.0, 112.8, 99.4, 27.1. MS (APCI) m/z [M+1]+ 270.2. 4.4.3 X-ray crystallographic analysis of compound 18 Crystals of [Z]-3-[Hydroxy-(4-phenoxy-phenyl)-methylene]-6-chloro-1,3-dihydro- indol-2-one (compound 18) were grown in solvent mixture of methanol and acetone and mounted on glass fibers. X-ray data were collected with a Bruker AXS SMART APEX diffractometer, using Mo Kα radiation at 223K, with the SMART suite of Programs (SMART version 5.628 (2003), Bruker AXS Inc., Madison, WI). Data were processed and corrected for Lorentz and polarization effects with SAINT (SAINT + version 6.22a (2001) Bruker AXS Inc., Madison, WI), and for absorption effect with SADABS (SADABS, version 2.10 (2001), University of Göttingen). Structural solution and refinement were carried out with the SHELXTL, suite of programs (SHELXTL, version 6.14 (2000), Bruker AXS Inc., Madison, WI). The structure was solved by direct methods to locate the heavy atoms, followed by difference maps for the light, non-hydrogen atoms. All non-hydrogen atoms were generally given anisotropic displacement parameters in the final model whereas H-atoms were placed at calculated positions. 4.5 Bioactivity investigations 4.5.1 DPPH free radical scavenging test 164 The measurement of antioxidant activity was performed according to a procedure described previously with minor modification 124. Quercetine was used as the standard antioxidant sample. DPPH and EtOH were used as stable free radical agent and blank, respectively. The sample stock solutions were prepared in DMSO at concentration of 100 mmol, and subsequently diluted by EtOH to the test concentrations. 20 µL of each compound at five concentrations (10-100 µM) was transferred to the vials in the 96-well plates which contained DPPH solution (0.2 mg/mL, 180 µL/vial). The reaction was kept in the dark for hrs. The antioxidant activity was measured as the decrease of the absorbance of DPPH and expressed as the percentage of the absorbance of a control DPPH solution with the addition of EtOH 20 µL. 4.5.2 Determination of total free phenolic compounds by Folin-Ciocalteu test The amount of total phenolic compounds in the samples was determined using FolinCiocalteu method125. Briefly, a calibration curve was prepared from gallic acid solution with concentration ranged from 0.03-0.09 mg/mL. In this method, mL distilled water was added into a 10 mL volumetric flask. Samples each mL with average concentrations of 1.25 mg/mL was transferred into the volumetric flask to obtain absorbance in the range of the prepared calibration curve. 0.2 mL of FolinCiocalteu reagent was added and mixed well for oxidation. After min, the reaction was neutralized with 0.4 mL satd. sodium carbonate (Na2CO3) solution. The solution is mixed well and made up to volume with distilled water. After a hr reaction in the dark, the absorbance was measured at 725 nm using Shimadzu UV-1601 UV-Visible Spectrophotometer. The results were expressed as gram of gallic acid equivalent (GAE) per 100 g of dry weight (DW) of the extracts 126. 4.5.3 MES and Rotorod test 165 Animals and drug administration Swiss albino mice (male, 20-30 g) were obtained from University Laboratory Animal Holding Unit at least two days prior to experimentation. They were kept under a natural light-dark cycle with food and water available. All animal experiments under the project No. R-148-000-049-112 were formally approved by the NUS institutional animal care and use committee (IACUC). Test samples were dissolved in dimethylsulfoxide (DMSO) and administered by intraperitoneal injection at mL/kg body weight. Mice were then subjected to Rotorod and MES tests for pharmacological activities 15-30 after injection. Prior to drug administration, the mice were put on an accelerating rotating treadmill which accelerated from 2-20 rotations per in min, and those that could pass this test by staying on the rotating rod for more than of the testing period were considered qualified for the following tests. Rotorod test 20 after drug administration, mice were put on the rotating rod, and the time duration when the mice could stand on the accelerating treadmill was recorded and used as a measure of impact on motor coordination. MES test Mice were shocked electrically with auricle electrodes at 40 mA for 0.2 sec. At his magnitude, all control (vehicle-injected) mice exhibited characteristic clonic-tonic convulsion which can be briefly described by four stages: clonus, tonic flexor, full tonic extensor (hindlimbs), and a pre-recovery period with loss of righting reflex. Under the influence of a drug with anticonvulsant activity, mice may show seizure 166 activities up to different stages depending on the dose and the efficacy of the drug used. Therefore, mice that showed the full range of seizure activities were arbitrarily assigned an “anticonvulsant score” of 1, while mice with seizure up to the flexor phase, 2; clonus, 3; and no seizure, 4. 4.5.4 Cell culture and cell viability test SH-SY5Y cell culture and experimental treatment of hydrogen peroxide Human neuroblastoma SH-SY5Y cells were obtained from American Type Culture Collection (Rockville, MD, USA). Cells were routinely cultured in MEM-Eagle/F-12 (HAM) (1:1), containing 10% FBS and a mixture of 2.5% sodium bicarbonate. Cell cultures were incubated at 37℃ in a humid 5% CO2-95% air environment. For experiments with H2O2, SH-SY5Y cells (6x103/well) were seeded in the cultured medium and allowed to attach for 24 hrs before drug treatment. Test samples were prepared at 100 µM in DMSO as stock. The sequential dilution using culture medium was to attain the required concentration. Test samples were added to the culture medium hrs before H2O2 treatment and cells were further incubated for 20 hrs. Vehicle-treated cultures were used as controls. Experimental treatment of serum deprivation SH-SY5Y cells were seeded (5x103/well) in 96-well plates and allowed to attach for 24 hrs before drug treatment. Serum-free medium was applied after removing medium in each well, and drugs dissolved in serum-free medium were given simultaneously. Cells were further incubated for 48 hrs. Vehicle-treated cultures were used as controls. Cell viability assay (MTT assay) 167 The MTT assay is based on the conversion of MTT to purple formazan crystals by viable cells. Briefly, after treatment the medium was removed and replaced with a fresh medium (100 µL/well). 10 µL of MTT (5 mg/mL) was added to each well. After incubation at 37 ℃ for hrs, the medium was removed and replaced with 100 µL DMSO. Absorbance was measured at 570 nm using a Tecan® microplate reader (Switzerland) after automatic subtraction of background readings. Cell viability was expressed as a percentage of control, untreated cells. 4.5.5 Electrophysiological study of compound 20 (3112) on Xenopus oocytes expressing NR1/NR2B cRNA for rat NR1-1a (hereafter NR1) and NR2B were synthesized in vitro and injected (5-10 ng) into stage V-VI Xenopus laevis oocytes, isolated as previously described127. Two electrode voltage-clamp current recordings were made 2-7 days post injection. The recording solution contained (in mmol) 90 NaCl, KCl, 10 HEPES, 0.5 BaCl2, 0.01 EDTA, 0.05 glycine (23 ℃); pH was adjusted to 7.3 with NaOH. EDTA (10 µM) was added to chelate contaminant extracellular divalent ions, including trace amounts of Zn2+. Solution exchange was computer controlled through an 8-modular valve positioned (Digital MVP Valve, Hamilton Company, Reno, NV). Voltage and current electrodes were filled with 0.3 and 3.0 M KCl, respectively, and current responses recorded at a holding potential of -50 mV. Data acquisition and voltage control were accomplished with a two-electrode voltage-clamp amplifier (OC-725, Warner Instruments, Hamden, CT). Current response was expressed as the percentage of maximal of induced current in the same oocyte and pooled among oocytes. 4.5.6 Statistics 168 ED50 values were calculated using Chou’s median-effect equation84. The seizure score obtained in the MES test were statistically evaluated using Kruskal-Wallis nonparametric ANOVA. In other cases, parametric ANOVA followed by Dunnett’s test was performed to compare the effects of drug treatment with the control. Difference between control and treatment would be considered statistically significant if P value was less than 0.05. All statistical tests were performed using commercially available SPSS 11.0 for Windows. 169 5. Bibliography 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. 16. 17. Phillipson, J. D.; Hemingway, S. R.; Ridsdale, C. E., Alkaloids of Uncaria, Part V. Their occurrence and chemotaxonomy. Lloydia 1978, 41, 503-570. Taguchi, H.; Yosioka, I.; Yamasaki, K.; Kim, I., Studies on the Constituents of Gastrodia elata Blume. Chemical & Pharmaceutical Bulletin 1981, 29 (1), 55-62. Hsieh, C.-L.; Tang, N.-Y.; Chiang, S.-Y.; Hsieh, C.-T.; Lin, J.-G., Anticonvulsive and free radical scavenging actions of two herbs, Uncaria rhynchophylla (Miq) jack and Gastrodia elata Bl., in kainic acid-treated rats. Life Sciences 1999, 65 (20), 2071-2082. (a) Kwan, P.; Brodie, M. J., Combination Therapy in Epilepsy: When and What to Use. Drugs 2006, 66 (14), 1817-1829; (b) Philip , N. P.; Walter, F.; Francesco, P.; Clementina , M. van R., The Importance of Drug Interactions in Epilepsy Therapy. Epilepsia 2002, 43 (4), 365-385. Firn, R. D.; Jones, C. G., Natural products - a simple model to explain chemical diversity. Natural Product Reports 2003, 20 (4), 382-391. Koehn, F. E.; Carter, G. T., The evolving role of natural products in drug discovery. Nature Reviews Drug Discovery 2005, (3), 206-220. Wagenaar, M., Pre-fractionated Microbial Samples-The Second Generation Natural Products Library at Wyeth. Molecules 2008, 13 (6), 1406-1426. Newman, D. J.; Cragg, G. M., Natural Products as Sources of New Drugs over the Last 25 Years. Journal of Natural Products 2007, 70 (3), 461-477. Paterson, I.; Anderson, E. A., The Renaissance of Natural Products as Drug Candidates. Science 2005, 310 (5747), 451-453. Bindseil, K. U.; Jakupovic, J.; Wolf, D.; Lavayre, J.; Leboul, J.; van der Pyl, D., Pure compound libraries; a new perspective for natural product based drug discovery. Drug Discovery Today 2001, (16), 840-847. Eldridge, G. R.; Vervoort, H. C.; Lee, C. M.; Cremin, P. A.; Williams, C. T.; Hart, S. M.; Goering, M. G.; O'Neil-Johnso, M.; Zeng, L., High-Throughput Method for the Production and Analysis of Large Natural Product Libraries for Drug Discovery. Analytical Chemistry 2002, 74 (16), 3963-3971. Suganuma, M.; Okabe, S.; Kai, Y.; Sueoka, N.; Sueoka, E.; Fujiki, H., Synergistic Effects of (-)-Epigallocatechin Gallate with (-)-Epicatechin, Sulindac, or Tamoxifen on Cancer-preventive Activity in the Human Lung CancerCell Line PC-9. Cancer Research 1999, 59 (1), 44-47. Khafif, A.; Schantz, S. P.; Chou, T.-C.; Edelstein, D.; Sacks, P. G., Quantitative chemopreventive synergism between (-)-epigallocatechin-3-gallate and curcumin in normal, premaligant and malignant human oral epithelial cells. Carcinogenesis 1998, 19 (3), 419-424. (a) Stermitz, F. R.; Lorenz, P.; Tawara, J. N.; Zenewicz, L. A.; Lewis, K., Synergy in a medicinal plant: Antimicrobial action of berberine potentiated by 5'methoxyhydnocarpin, a multidrug pump inhibitor. Proceedings of the National Academy of Sciences of the United States of America 2000, 97 (4), 1433-1437; (b) Stermitz, F. R.; Tawara-Matsuda, J.; Lorenz, P.; Mueller, P.; Zenewicz, L.; Lewis, K., 5'-Methoxyhydnocarpin-D and Pheophorbide-A: Berberis Species Components that Potentiate Berberine Growth Inhibition of Resistant Staphylococcus aureus. Journal of Natural Products 2000, 63 (8), 1146-1149. Lin, F.-M.; Chen, L.-R.; Lin, E.-H.; Ke, F.-C.; Chen, H.-Y.; Tsai, M.-J.; Hsiao, P.-W., Compounds from Wedelia chinensis synergistically suppress androgen activity and growth in prostate cancer cells. Carcinogenesis 2007, 28 (12), 2521-2529. Pappa, G.; Strathmann, J.; Löwinger, M.; Bartsch, H.; Gerhäuser, C., Quantitative combination effects between sulforaphane and 3,3'-diindolylmethane on proliferation of human colon cancer cells in vitro. Carcinogenesis 2007, 28 (7), 1471-1477. Sauer, U.; Heinemann, M.; Zamboni, N., Getting Closer to the Whole Picture. Science 2007, 316 (5824), 550-551. 170 18. 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. (a) Borisy, A. A.; Elliott, P. J.; Hurst, N. W.; Lee, M. S.; Lehar, J.; Price, E. R.; Serbedzija, G.; Zimmermann, G. R.; Foley, M. A.; Stockwell, B. R.; Keith, C. T., Systematic Discovery of Multicomponent Therapeutics. Proceedings of the National Academy of Sciences of the United States of America 2003, 100 (13), 7977-7982; (b) Fitzgerald, J. B.; Shoeberl, B.; Nielsen, U. B.; Sorger, P. K., Systems Biology and Combination Therapy in the Quest for Clinical Efficacy. Nature Chemical Biology 2006, (9), 458-466. (a) Rochfort, S., Metabolomics Reviewed: A New "Omic" Platform Technology for Systems Biology and Implications for Natural Products Research. Journal of Natural Products 2005, 68 (12), 1813-1820; (b) Wang, M.; Lamers, R.-J. A. N.; Korthout, H. A. A. J.; van Nesselrooij, J., H. J.; Witcamp, R. F.; van der Heijden, R.; Voshol, P. J.; Havekes, L. M.; Verpoorte, R.; Van der Greef, J., Metabolomics in the Context of Systems Biology: Bridging Traditional Chinese Medicine and Molecular Pharmacology. Phytotherapy Research 2005, 19, 173-182. Wang, Y.; Tang, H.; Nicholson, J. K.; Hylands, P. J.; Sampson, J.; Holmes, E., A Metabonomic Strategy for the Detection of the Metabolic Effects of Chamomile (Matricaria recutita L.) Ingestion. Journal of Agricultural and Food Chemistry 2005, 53 (2), 191-196. Chabala, J. C.; Mrozik, H.; Tolman, R. L.; Eskola, P.; Lusi, A.; Peterson, L. H.; Woods, M. F.; Fisher, M. H.; Campbell, W. C., Ivermectin, a new broad-spectrum antiparasitic agent. Journal of Medicinal Chemistry 1980, 23 (10), 1134-1136. He, H.; Williamson, R. T.; Shen, B.; Graziani, E. I.; Yang, H. Y.; Sakya, S. M.; Petersen, P. J.; Carter, G. T., Mannopeptimycins, Novel Antibacterial Glycopeptides from Streptomyces hygroscopicus, LL-AC98. Journal of the American Chemical Society 2002, 124 (33), 9729-9736. Atoui, A. K.; Mansouri, A.; Boskou, G.; Kefalas, P., Tea and herbal infusions: Their antioxidant activity and phenolic profile. Food Chemistry 2005, 89 (1), 27-36. Lee, J.; Son, D.; Lee, P.; Kim, S.-Y.; Kim, H.; Kim, C.-J.; Lim, E., Alkaloid fraction of Uncaria rhynchophylla protects against N-methyl--aspartate-induced apoptosis in rat hippocampal slices. Neuroscience Letters 2003, 348 (1), 51-55. van Beek, T. A., Chemical analysis of Ginkgo biloba leaves and extracts. Journal of Chromatography A 2002, 967 (1), 21-55. A.-L. Cogne, E. F. Q., J.-L. Wolfender, A. Marston, S. Mavi, K. Hostettmann,, On-line identification of unstable catalpol derivatives from Jamesbrittenia fodina by LC-MS and LC-NMR. Phytochemical Analysis 2003, 14 (2), 67-73. Kang, S. W.; Kim, M. C.; Kim, C. Y.; Jung, S. H.; Um, B. H., The Rapid Identification of Isoflavonoids from Belamcanda chinensis by LC-NMR and LC-MS. Chemical & Pharmaceutical Bulletin 2008, 56 (10), 1452-1454. Iwasa, K.; Takahashi, T.; Nishiyama, Y.; Moriyasu, M.; Sugiura, M.; Takeuchi, A.; Tode, C.; Tokuda, H.; Takeda, K., Online Structural Elucidation of Alkaloids and Other Constituents in Crude Extracts and Cultured Cells of Nandina domestica by Combination of LC-MS/MS, LC-NMR, and LC-CD Analyses. Journal of Natural Products 2008, 71 (8), 1376-1385. (a) Martínez Vidal, J. L.; Belmonte Vega, A.; Garrido Frenich, A.; Egea González, F. J.; Arrebola Liebanas, F. J., Determination of fifteen priority phenolic compounds in environmental samples from Andalusia (Spain) by liquid chromatography–mass spectrometry. Analytical and Bioanalytical Chemistry 2004, 379 (1), 125-130; (b) Petsalo, A.; Jalonen, J.; Tolonen, A., Identification of flavonoids of Rhodiola rosea by liquid chromatography-tandem mass spectrometry. Journal of Chromatography A 2006, 1112 (1-2), 224-231; (c) Sánchez-Rabaneda, F.; Jáuregui, O.; Casals, I.; AndrésLacueva, C.; Izquierdo-Pulido, M.; Lamuela-Raventós, R. M., Liquid chromatographic/electrospray ionization tandem mass spectrometric study of the phenolic composition of cocoa (Theobroma cacao). Journal of Mass Spectrometry 2003, 38 (1), 35-42. Pharmacopoeia of the People's Republic of China. 1995; p 45-46. 171 31. 32. 33. 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. (a) Heitzman, M. E.; Neto, C. C.; Winiarz, E.; Vaisberg, A. J.; Hammond, G. B., Ethnobotany, phytochemistry and pharmacology of Uncaria (Rubiaceae). Phytochemistry 2005, 66 (1), 5-29; (b) Gerhard, L., Advances in chemistry and bioactivity of the genus Uncaria. Phytotherapy Research 2004, 18 (4), 259-274. Kuramochi, T.; Chu, J.; Suga, T., Gou-teng (from Miquel)-induced endotheliumdependent and -independent relaxations in the isolated rat aorta. Life Sciences 1994, 54 (26), 2061-2069. Suk, K.; Kim, S. Y.; Leem, K.; Kim, Y. O.; Park, S. Y.; Hur, J.; Baek, J.; Lee, K. J.; Zheng, H. Z.; Kim, H., Neuroprotection by methanol extract of Uncaria rhynchophylla against global cerebral ischemia in rats. Life Sciences 2002, 70 (21), 2467-2480. Liu, J.; Mori, A., Antioxidant and free radical scavenging activities of Gastrodia elata Bl. and Uncaria rhynchophylla (Miq.) Jacks. Neuropharmacology 1992, 31 (12), 12871298. (a) Wurm, M.; Kacani, L.; Laus, G.; Keplinger, K.; Dierich, M., Pentacyclic Oxindole Alkaloids from Uncaria tomentosa Induce Human Endothelial Cells to Release a Lymphocyte-Proliferation-Regulating Factor. Planta Medica 1998, (8), 701-704; (b) Winkler, C.; Wirleitner, B.; Schroecksnadel, K.; Schennach, H.; Mur, E.; Fuchs, D., In vitro Effects of Two Extracts and Two Pure Alkaloid Preparations of Uncaria tomentosa on Peripheral Blood Mononuclear Cells. Planta Medica 2004, (3), 205-210. Laus, G., Kinetics of isomerization of tetracyclic spiro oxindole alkaloids. Journal of the Chemical Society 1998, (2), 315-318. (a) Horie, S.; Yano, S.; Aimi, N.; Sakai, S.-i.; Watanabe, K., Effects of hirsutine, an antihypertensive indole alkaloid from Uncaria rhynchophylla, on intracellular calcium in rat thoracic aorta. Life Sciences 1992, 50 (7), 491-498; (b) Masumiya, H.; Saitoh, T.; Tanaka, Y.; Horie, S.; Aimi, N.; Takayama, H.; Tanaka, H.; Shigenobu, K., Effects of hirsutine and dihydrocorynantheine on the action potentials of sino-atrial node, atrium and ventricle. Life Sciences 1999, 65 (22), 2333-2341. Yuzurihara, M.; Ikarashi, Y.; Goto, K.; Sakakibara, I.; Hayakawa, T.; Sasaki, H., Geissoschizine methyl ether, an indole alkaloid extracted from Uncariae Ramulus et Uncus, is a potent vasorelaxant of isolated rat aorta. European Journal of Pharmacology 2002, 444 (3), 183-189. Stæk, D.; Lemmich, E.; Christensen, J.; Kharazmi, A.; Olsen, C. E.; Jaroszewski, J., Leishmanicidal, Antiplasmodial and Cytotoxic Activity of Indole Alkaloids from Corynanthe pachyceras. Planta Medica 2000, (6), 531-536. Kang, T.-H.; Matsumoto, K.; Tohda, M.; Murakami, Y.; Takayama, H.; Kitajima, M.; Aimi, N.; Watanabe, H., Pteropodine and isopteropodine positively modulate the function of rat muscarinic M1 and 5-HT2 receptors expressed in Xenopus oocyte. European Journal of Pharmacology 2002, 444 (1-2), 39-45. (a) Kang, T.-H.; Murakami, Y.; Matsumoto, K.; Takayama, H.; Kitajima, M.; Aimi, N.; Watanabe, H., Rhynchophylline and isorhynchophylline inhibit NMDA receptors expressed in Xenopus oocytes. European Journal of Pharmacology 2002, 455 (1), 2734; (b) Kang, T.-H.; Murakami, Y.; Takayama, H.; Kitajima, M.; Aimi, N.; Watanabe, H.; Matsumoto, K., Protective effect of rhynchophylline and isorhynchophylline on in vitro ischemia-induced neuronal damage in the hippocampus: putative neurotransmitter receptors involved in their action. Life Sciences 2004, 76 (3), 331-343; (c) Matsumoto, K.; Morishige, R.; Murakami, Y.; Tohda, M.; Takayama, H.; Sakakibara, I.; Watanabe, H., Suppressive effects of isorhynchophylline on 5-HT2A receptor function in the brain: Behavioural and electrophysiological studies. European Journal of Pharmacology 2005, 517 (3), 191-199. SHI Jing-Shan, Y. J.-X., CHEN Xiu-Ping, XIU Rui-Xia, Pharmacological actions of Uncaria alkaloids, rhynchophylline and isorhynchophylline. Acta Pharmacologica Sinica 2003, 24 (2), 97-101. White, H. S. P., Animal models of epileptogenesis. Neurology 2002, 59(9) Supplement (5), S7-S14. 172 44. 45. 46. 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. Mohamed, A. F. A. F.; Matsumoto, K.; Tabata, K.; Takayama, H.; Kitajima, M.; Aimi, N.; Watanabe, H., Effects of Uncaria tomentosa Total Alkaloid and its Components on Experimental Amnesia in Mice: Elucidation Using the Passive Avoidance Test. Journal of Pharmacy and Pharmacology 2000, 52, 1553-1561. (a) Lee, J. S.; Kim, J.; Kim, B. Y.; Lee, H. S.; Ahn, J. S.; Chang, Y. S., Inhibition of Phospholipase Cγ1 and Cancer Cell Proliferation by Triterpene Esters from Uncaria rhynchophylla. Journal of Natural Products 2000, 63 (6), 753-756; (b) Lee, J. S.; Yang, M. Y.; Yeo, H.; Kim, J.; Lee, H. S.; Ahn, J. S., Uncarinic acids: Phospholipase Cγ1 inhibitors from hooks of Uncaria rhynchophylla. Bioorganic & Medicinal Chemistry Letters 1999, (10), 1429-1432. Sekiya, N.; Shimada, Y.; Shibahara, N.; Takagi, S.; Yokoyama, K.; Kasahara, Y.; Sakakibara, I.; Terasawa, K., Inhibitory effects of Choto-san (Diao-teng-san), and hooks and stems of Uncaria sinensis on free radical-induced lysis of rat red blood cells. Phytomedicine 2002, (7), 636-640. Law, K. H.; Das, N. P., Studies on the Formation and Growth of Uncaria elliptica Tissue Culture. Journal of Natural Products 1990, 53 (1), 125-130. Ojemann, L. M.; Nelson, W. L.; Shin, D. S.; Rowe, A. O.; Buchanan, R. A., Tian ma, an ancient Chinese herb, offers new options for the treatment of epilepsy and other conditions. Epilepsy & Behavior 2006, (2), 376-383. Huang, N.-K.; Lin, Y.-L.; Cheng, J.-J.; Lai, W.-L., Gastrodia elata prevents rat pheochromocytoma cells from serum-deprived apoptosis: the role of the MAPK family. Life Sciences 2004, 75 (13), 1649-1657. (a) Kim, H.-J.; Lee, S.-R.; Moon, K.-D., Ether fraction of methanol extracts of Gastrodia elata, medicinal herb protects against neuronal cell damage after transient global ischemia in gerbils. Phytotherapy Research 2003, 17 (8), 909-912; (b) Kim, H.J.; Moon, K.-D.; Lee, D.-S.; Lee, S.-H., Ethyl ether fraction of Gastrodia elata Blume protects amyloid β peptide-induced cell death. Journal of Ethnopharmacology 2003, 84 (1), 95-98; (c) Kim, H.-J.; Moon, K.-D.; Oh, S.-Y.; Kim, S.-P.; Lee, S.-R., Ether fraction of methanol extracts of Gastrodia elata, a traditional medicinal herb, protects against kainic acid-induced neuronal damage in the mouse hippocampus. Neuroscience Letters 2001, 314 (1-2), 65-68. Yu, S. J.; Kim, J. R.; Lee, C. K.; Han, J. E.; Lee, J. H.; Kim, H.-S.; Hong, J. H.; Kang, S. G., Gastrodia elata Blume and an Active Component, p-Hydroxybenzyl Alcohol Reduce Focal Ischemic Brain Injury through Antioxidant Related Gene Expressions. Biological & Pharmaceutical Bulletin 2005, 28 (6), 1016-1020. Zeng, X.; Zhang, S.; Zhang, L.; Zhang, K.; Zheng, X., A Study of the Neuroprotective Effect of the Phenolic Glucoside Gastrodin during Cerebral Ischemia in vivo and in vitro. Planta Medica 2006, (15), 1359-1365. Wu, C.-R.; Hsieh, M.-T.; Huang, S.-C.; Peng, W.-H.; Chang, Y.-S.; Chen, C.-F., Effects of Gastrodia elata and its Active Constituents on Scopolamine-Induced Amnesia in Rats. Planta Medica 1996, (4), 317-321. An, S.-J.; Park, S.-K.; Hwang, I. K.; Choi, S. Y.; Kim, S. K.; Kwon, O.-S.; Jung, S. J.; Baek, N.-I.; Lee, H. Y.; Won, M. H.; Kang, T.-C., Gastrodin decreases immunoreactivities of γ-aminobutyric acid shunt enzymes in the hippocampus of seizure-sensitive gerbils. Journal of Neuroscience Research 2003, 71 (4), 534-543. Ha, J.-H.; Lee, D.-U.; Lee, J.-T.; Kim, J.-S.; Yong, C.-S.; Kim, J.-A.; Ha, J.-S.; Huh, K., 4-Hydroxybenzaldehyde from Gastrodia elata B1. is active in the antioxidation and GABAergic neuromodulation of the rat brain. Journal of Ethnopharmacology 2000, 73 (1-2), 329-333. Hayashi, J.; Sekine, T.; Deguchi, S.; Lin, Q.; Horie, S.; Tsuchiya, S.; Yano, S.; Watanabe, K.; Ikegami, F., Phenolic compounds from Gastrodia rhizome and relaxant effects of related compounds on isolated smooth muscle preparation. Phytochemistry 2002, 59 (5), 513-519. 173 57. 58. 59. 60. 61. 62. 63. 64. 65. 66. 67. 68. 69. 70. Lee, Y.-K.; Woo, M.-H.; Kim, C.-H.; Kim, Y.; Lee, S.-H.; Jeong, B.-S.; Chang, H.-W.; Son, J.-K., Two New Benzofurans from Gastrodia elata and their DNA Topoisomerases I and II Inhibitory Activities. Planta Medica 2007, 73 (12), 1287-1291. Huang, N.-K.; Chern, Y.; Fang, J.-M.; Lin, C.-I.; Chen, W.-P.; Lin, Y.-L., Neuroprotective Principles from Gastrodia elata. Journal of Natural Products 2007, 70 (4), 571-574. Cole, A. J.; Dichter, M., Neuroprotection and antiepileptogenesis: Overview, definitions, and context. Neurology 2002, 59 (9, supplement 5), 2. White, H. S., Preclinical Development of Antiepileptic Drugs: Past, Present, and Future Directions. Epilepsia 2003, 44 (s7), 2-8. (a) Wuarin, J.-P.; Dudek, F. E., Electrographic Seizures and New Recurrent Excitatory Circuits in the Dentate Gyrus of Hippocampal Slices from Kainate-Treated Epileptic Rats. The Journal of Neuroscience 1996, 16 (14), 4438-4448; (b) Gibbs, J. W., III; Shumate, M. D.; Coulter, D. A., Differential Epilepsy-Associated Alterations in Postsynaptic GABAA Receptor Function in Dentate Granule and CA1 Neurons. Journal of Neurophysiology 1997, 77 (4), 1924-1938. DeLorenzo, R. J.; Sun, D. A.; Deshpande, L. S., Cellular mechanisms underlying acquired epilepsy: The calcium hypothesis of the induction and maintainance of epilepsy. Pharmacology & Therapeutics 2005, 105 (3), 229-266. Holtkamp, M.; Meierkord, H., Anticonvulsant, antiepileptogenic, and antiictogenic pharmacostrategies. Cellular and Molecular Life Sciences (CMLS) 2007, 64 (15), 2023-2041. Cole, A. J. M. D. F.; Dichter, M. M. D. P., Neuroprotection and antiepileptogenesis: Overview, definitions, and context. Neurology 2002, 59(9) Supplement (5), S1-S2. Fisher, R. S., Animal models of the epilepsies. Brain Research Reviews 1989, 14 (3), 245-278. Pitkanen, A., Drug-mediated neuroprotection and antiepileptogenesis: Animal data. Neurology 2002, 59(9) Supplement (5), 27S-33. (a) O'Neil, M. J.; Bogaert, L.; Hicks, C. A.; Bond, A.; Ward, M. A.; Ebinger, G.; Ornstein, P. L.; Michotte, Y.; Lodge, D., LY377770, a Novel iGlu5 Kainate Receptor Antagonist with Neuroprotective Effects in Global and Focal Cerebral Ischemia. Neuropharmacology 2000, 39, 1575-1588; (b) Stepien, K.; Tomaszewski, M.; Czuczwar, S. J., Profile of Anticonvulsant Activity and Neuroprotective Effects of Novel and Potential Antiepileptic Drugs - An Update. Pharmacological Reports 2005, 57, 719-733. (a) Arthur, P. G.; Lim, S. C. C.; Meloni, B. P.; Munns, S. E.; Chan, A.; Knuckey, N. W., The Protective Effect of Hypoxic Preconditioning on Cortical Neuronal Cultures is Associated with Increases in the Activity of Several Antioxidant Enzymes. Brain Research 2004, 1017, 146-154; (b) Munns, S. E.; Meloni, B. P.; Knuckey, N. W.; Arthur, P. G., Primary Cortical Neuronal Cultures Reduce Cellular Energy Utilization during Anoxic Energy Deprivation. Journal of Neurochemistry 2003, 87, 764-772. (a) Maher, P.; Salgado, K. F.; Zivin, J. A.; Lapchak, P. A., A novel approach to screening for new neuroprotective compounds for the treatment of stroke. Brain Research 2007, 1173, 117-125; (b) Kang, S. S.; Lee, J. Y.; Choi, Y. K.; Kim, G. S.; Han, B. H., Neuroprotective effects of flavones on hydrogen peroxide-induced apoptosis in SH-SY5Y neuroblastoma cells. Bioorganic & Medicinal Chemistry Letters 2004, 14 (9), 2261-2264; (c) Gao, M.; Zhang, W.-c.; Liu, Q.-s.; Hu, J.-j.; Liu, G.-t.; Du, G.-h., Pinocembrin Prevents Glutamate-induced Apoptosis in SH-SY5Y Neuronal Cells via decrease of bax/bcl-2 ratio. European Journal of Pharmacology 2008, 591, 73-79; (d) Ji, Z. N.; Dong, T. T. X.; Ye, W. C.; Choi, R. C.; Lo, C. K.; Tsim, K. W. K., Ginsenoside Re Attenuate β-amyloid and Serum-free induced Neurotoxicity in PC12 Cells. Journal of Ethnopharmacology 2006, 107, 48-52. Loscher, W.; Schmidt, D., Strategies in antiepileptic drug development: is rational drug design superior to random screening and structural variation? Epilepsy Research 1994, 17 (2), 95-134. 174 71. 72. 73. 74. 75. 76. 77. 78. 79. 80. 81. 82. 83. 84. 85. Kemp, J. A.; Leeson, P. D., The glycine site of the NMDA receptor -- five years on. Trends in Pharmacological Sciences 1993, 14 (1), 20-25. Wong, E. H.; Kemp, J. A.; Priestley, T.; Knight, A. R.; Woodruff, G. N.; Iversen, L. L., The Anticonvulsant MK-801 is a Potent N-methyl-D-aspartate antagonist. Proceedings of the National Academy of Sciences of the United States of America 1986, 83 (18), 7104-7108. Stasheff, S. F. A., William W.; Clark, Suzanne; Wilson, Wilkie A., NMDA Antagonists Differentiate Epileptogenesis from Seizure Expression in an in vitro Model. Science 1989, 245 (4918), 4. Palmer, G. C., Neuroprotection by NMDA Receptor Antagonists in a Variety of Neuropathologies. Current Drug Targets 2001, 2, 241-271. Catarzi, D.; Colotta, V.; Varano, F., Competitive Gly/NMDA Receptor Antagonists. Current Topics in Medicinal Chemistry 2006, 6, 809-821. (a) Johnson, J. W.; Ascher, P., Glycine potentiates the NMDA response in cultured mouse brain neurons. Nature 1987, 325 (6104), 529-531; (b) Kleckner, N. W.; Dingledine, R., Requirement for Glycine in Activation of NMDA-Receptors Expressed in Xenopus Oocytes. Science 1988, 241 (4867), 835-837. Brauner-Osborne, H.; Egebjerg, J.; Nielsen, E. O.; Madsen, U.; Krogsgaard-Larsen, P., Ligands for Glutamate Receptors: Design and Therapeutic Prospects. Journal of Medicinal Chemistry 2000, 43 (14), 2609-2645. (a) Klimaviciusa, L.; Safiulina, D.; Kaasik, A.; Klusa, V.; Zharkovsky, A., The effects of glutamate receptor antagonists on cerebellar granule cell survival and development. NeuroToxicology 2008, 29 (1), 101-108; (b) Priestley, T.; Horne, A. L.; McKernan, R. M.; Kemp, J. A., The effect of NMDA receptor glycine site antagonists on hypoxiainduced neurodegeneration of rat cortical cell cultures. Brain Research 1990, 531 (1-2), 183-188. Danysz, W.; Parsons, C. G., Glycine and N-Methyl-D-Aspartate Receptors: Physiological Significance and Possible Therapeutic Applications. Pharmacological Reviews 1998, 50 (4), 597-664. Furukawa, H.; Gouaux, E., Mechanisms of activation, inhibition and specificity: crystal structures of the NMDA receptor NR1 ligand-binding core. The EMBO Journal 2003, 22 (12), 2873-2885. Leeson, P. D.; Iversen, L. L., The Glycine Site on the NMDA Receptor: StructureActivity Relationships and Therapeutic Potential. Journal of Medicinal Chemistry 1994, 37 (24), 4053-4067. (a) Leeson, P. D.; Baker, R.; Carling, R. W.; Kulagowski, J. J.; Mawer, I. M.; Ridgill, M. P.; Rowley, M.; Smith, J. D.; Stansfield, I.; Stevenson, G. I.; Foster, A. C.; Kemp, J. A., Amino acid bioisosteres: design of 2-quinolone derivatives as glycine-site Nmethyl-D-aspartate receptor antagonists. Bioorganic & Medicinal Chemistry Letters 1993, (2), 299-304; (b) McQuaid, L. A.; Smith, E. C. R.; Lodge, D.; Pralong, E.; Wikel, J. H.; Calligaro, D. O.; O'Malley, P. J., 3-Phenyl-4-hydroxyquinolin-2(1H)-ones: potent and selective antagonists at the strychnine-insensitive glycine site on the Nmethyl-D-aspartate receptor complex. Journal of Medicinal Chemistry 1992, 35 (18), 3423-3425. (a) Kadin, S. B. 1,3-disubstituted 2-oxindoles as analgesic and anti-inflammatory agents. 4,721,712, 1988; (b) Kadin, S. B. 3-substituted 2-oxindole-1-carboxamides as analgesic and anti-inflammatory agents. 4,556,672, 1985; (c) Kadin, S. B. Analgesic and antiinflammatory 1,3-diacyl-2-oxindole compounds. 4,690,943, 1987; (d) Kadin, S. B. N,3-disubstituted 2-oxindole-1-carboxamides as analgesic and antiinflammatory agents. 4,569,942, 1986. Chou, T.-C., Theoretical Basis, Experimental Design, and Computerized Simulation of Synergism and Antagonism in Drug Combination Studies. Pharmacological Reviews 2006, 58 (3), 621-681. Borowicz, K. K.; Kimber-Trojnar, Z.; Ratnaraj, N.; Patsalos, P. N.; Luszczki, J. J.; Czuczwar, S. J., Isobolographic analysis of interactions between losigamone and 175 86. 87. 88. 89. 90. 91. 92. 93. 94. 95. 96. 97. 98. conventional antiepileptic drugs in the mouse maximal electroshock model. European Neuropsychopharmacology 2007, 17 (2), 94-101. David Phillipson, J.; Hemingway, S. R., Alkaloids of Uncaria attenuata, U. orientalis and U. Canescens. Phytochemistry 1975, 14 (8), 1855-1863. Lounasmaa, M.; Kan, S.-K., A 400 mhz 1H NMR study of the eight basic heteroyohimbine alkaloids. Tetrahedron 1980, 36 (11), 1607-1611. Wenkert, E.; Chang, C.-J.; Chawla, H. P. S.; Cochran, D. W.; Hagaman, E. W.; King, J. C.; Orito, K., General methods of synthesis of indole alkaloids. 14. Short routes of construction of yohimboid and ajmalicinoid alkaloid systems and their carbon-13 nuclear magnetic resonance spectral analysis. Journal of the American Chemical Society 1976, 98 (12), 3645-3655. (a) Trager, W. F.; Lee, C. M.; Phillipson, J. D.; Beckett, A. H., The absolute configuration of paynantheine and hirsutine. Tetrahedron 1967, 23 (2), 1043-1047; (b) Koch, M. C.; Plat, M. M.; Preaux, N.; Gottlieb, H. E.; Hagaman, E. W.; Schell, F. M.; Wenkert, E., Carbon-13 nuclear magnetic resonance spectroscopy of naturally occurring substances. XXXIII. Ochrolifuanines and emetine. The Journal of Organic Chemistry 1975, 40 (19), 2836-2838; (c) Wenkert, E.; Bindra, J. S.; Chang, C.-J.; Cochran, D. W.; Schell, F. M., Carbon-13 nuclear magnetic resonance spectroscopy of naturally occurring substances. Alkaloids. Accounts of Chemical Research 1974, (2), 46-51. Lounasmaa, M.; Jokela, R.; Laine, C.; Hanhinen, P., Short, Stereospecific Total Synthesis of (±)- Corynantheidine. Tetrahedron Letters 1995, 36 (47), 8687-8688. (a) Li, H.-X.; Ding, M.-Y.; Lv, K.; Yang, W.; Yu, J.-Y., Identification and determination of the active compounds in Gastrodia elata Blume by HPLC. Journal of Liquid Chromatography and Related Technologies 2001, 21 (4), 579-588; (b) Liu, C.; Liu, M.; Zhu, P., Determination of gastrodin,p-hydroxybenzyl alcohol, vanillyl alcohol,p-hydroxylbenzaldehyde and vanillin in tall gastrodia tuber by highperformance liquid chromatography. Chromatographia 2002, 55 (5), 317-320; (c) Ruan, D.; Yang, C.; Pu, X., The quantitative analysis of phenols from Gastrodia elata and its related plants by HPLC. Acta Botanica Yunnanica 1988, 10 (2), 231-237. Li, H. X.; Ding, M. Y.; Yu, J. Y., Simultaneous Determination of pHydroxybenzaldehyde, p-Hydroxybenzyl Alcohol, 4-(beta-D-Glucopyranosyloxy)Benzyl Alcohol, and Sugars in Gastrodia elata Blume Measured as Their Acetylated Derivatives by GC-MS. Journal of Chromatographic Science 2001, 39 (6), 251-254. Wang, L.; Xiao, H.; Hu, J.; Wang, J.; Liang, X., Study of p-hydroxy phenylic compounds in Gastrodia elata blume by high performance liquid chromatographyelectrospray-mass spectrometry. Chinese Journal of Analytical Chemistry 2003, 31 (8), 954-957. Wang, L.; Xiao, H.; Liang, X.; Wei, L., Identification of phenolics and nucleoside derivatives in Gastrodia elata by HPLC-UV-MS. Journal of Separation Science 2007, 30 (10), 1488-1495. Li, H. x.; Ding, M. y.; Yu, J. y., Simultaneous Determination of pHydroxybenzaldehyde, p-Hydroxybenzyl Alcohol, 4-(beta-D-Glucopyranosyloxy)Benzyl Alcohol, and Sugars in Gastrodia elata Blume Measured as Their Acetylated Derivatives by GC-MS. Journal of Chromatographic Science 2001, 39, 251-254. Kawada, A.; Yasuda, K.; Abe, H.; Harayama, T., Rare Earth Metal Trifluoromethanesulfonates Catalyzed Benzyl-Etherification. Chemical & Pharmaceutical Bulletin 2002, 50 (3), 380-383. Kim, S.; Chung, K. N.; Yang, S., Direct synthesis of ethers via zinc chloride-mediated etherification of alcohols in dichloroethane. The Journal of Organic Chemistry 1987, 52 (17), 3917-3919. Finn, S. R.; James, J. W., Formaldehyde Condensations with Phenol and Its Homologues, XVIII – chromatographic analysis. Journal of Applied Chemistry 1956, 10, 466. 176 99. 100. 101. 102. 103. 104. 105. 106. 107. 108. 109. 110. 111. 112. Wong, P. T.-H.; Tan, S.-F., Pharmacological Profiles of 5-Phenylmethylenehydantoin and Its Methoxy Substituted Derivatives. The Japanese Journal of Pharmacology 1989, 49 (3), 309-315. Barry, H., Oxidative stress and neurodegeneration: where are we now? Journal of Neurochemistry 2006, 97 (6), 1634-1658. (a) Yoshida, T.; Nakamura, H.; Masutani, H.; Yodoi, J., The involvement of thioredoxin and thioredoxin binding protein-2 on cellular proliferation and aging process. Annals of the New York Academy of Sciences 2005, 1055 (1), 1-12; (b) Papadia, S.; Soriano, F. X.; Lévéillé, F.; Martel, M.-A.; Dakin, K. A.; Hansen, H. H.; Kaindl, A.; Sifringer, M.; Fowler, J.; Stefovska, V.; Mckenzie, G.; Craigon, M.; Corriveau, R.; Ghazal, P.; Horsburgh, K.; Yankner, B. A.; Wyllie, D. J. A.; Ikonomidou, C.; Hardinghan, G. E., Synaptic NMDA receptor activity boosts intrinsic antioxidant defenses. Nature Neuroscience 2008, 11 (4), 476-487. Halliwell, B.; Gutteridge, J. M. C., Biologically relevant metal ion-dependent hydroxyl radical generation An update. FEBS Letters 1992, 307 (1), 108-112. Murphy, T. H.; Miyamoto, M.; Sastre, A.; Schnaar, R. L.; Coyle, J. T., Glutamate toxicity in a neuronal cell line involves inhibition of cystine transport leading to oxidative stress. Neuron 1989, (6), 1547-1558. Rukenstein, A.; Rydel, R.; Greene, L., Multiple agents rescue PC12 cells from serumfree cell death by translation- and transcription-independent mechanisms. The Journal of Neuroscience 1991, 11 (8), 2552-2563. Rydel, R. E.; Greene, L. A., cAMP analogs promote survival and neurite outgrowth in cultures of rat sympathetic and sensory neurons independently of nerve growth factor. Proceedings of the National Academy of Sciences of the United States of America 1988, 85 (4), 1257-1261. (a) Mizumaki, Y.; Kurimoto, M.; Hirashima, Y.; Nishijima, M.; Kamiyama, H.; Nagai, S.; Takaku, A.; Sugihara, K.; Shimizu, M.; Endo, S., Lipophilic fraction of Panax ginseng induces neuronal differentiation of PC12 cells and promotes neuronal survival of rat cortical neurons by protein kinase C dependent manner. Brain Research 2002, 950 (1-2), 254-260; (b) Yun, H.; Chou, T.-C.; Dong, H.; Tian, Y.; Li, Y.-m.; Danishefsky, S. J., Total Synthesis as a Resource in Drug Discovery: the First In Vivo Evaluation of Panaxytriol and Its Derivatives. The Journal of Organic Chemistry 2005, 70 (25), 10375-10380. Lee, S. R.; Kwak, J. H.; Kim, H. J.; Pyo, S., Neuroprotective Effects of Kobophenol A against the Withdrawas of Tropic Support, Nitrosative Stress, and Mitrochondria Damage in SH-SY5Y neuroblastoma cells. Bioorganic & Medicinal Chemistry Letters 2007, 17, 1879-1882. Prochaska, H. J.; De Long, M. J.; Talalay, P., On the mechanisms of induction of cancer-protective enzymes: a unifying proposal. Proceedings of the National Academy of Sciences of the United States of America 1985, 82 (23), 8232-8236. Talalay, P.; De Long, M. J.; Prochaska, H. J., Identification of a common chemical signal regulating the induction of enzymes that protect against chemical carcinogenesis. Proceedings of the National Academy of Sciences of the United States of America 1988, 85 (21), 8261-8265. Chew, E.-H.; Lu, J.; Bradshaw, T. D.; Holmgren, A., Thioredoxin reductase inhibition by antitumor quinols: a quinol pharmacophore effect correlating to antiproliferative activity. FASEB J. 2008, 22 (6), 2072-2083. Shimada, Y.; Goto, H.; Itoh, T.; Sakakibara, I.; Kubo, M.; Sasaki, H.; Terasawa, K., Evaluation of the Protective Effects of Alkaloids Isolated from the Hooks and Stems of Uncaria sinensis on Glutamate-induced Neuronal Death in Cultured Cerebellar Granule Cells from Rats. Journal of Pharmacy and Pharmacology 1999, 51, 715-722. (a) Na, M.; Kim, Y. H.; Min, B. S.; Bae, K.; Kamiryo, Y.; Senoo, Y.-i.; Yokoo, S.; Miwa, N.; Song, K. S.; Seong, Y. H., Cytoprotective effect on oxidative stress and inhibitory effect on cellular aging of Uncaria sinensis Havil. Journal of Ethnopharmacology 2004, 95 (2-3), 127-132; (b) Lee, J.; Son, D.; Lee, P.; Kim, D.-K.; 177 113. 114. 115. 116. 117. 118. 119. 120. 121. 122. 123. 124. 125. Shin, M.-C.; Jang, M.-H.; Kim, C.-J.; Kim, Y.-S.; Kim, S.-Y.; Kim, H., Protective Effect of Methanol Extract of Uncaria rhynchophylla Against Excitotoxicity Induced by N-Methyl-D-Aspartate in Rat Hippocampus. Journal of pharmacological Sciences 2003, 92 (1), 70-73. Kulagowski, J. J.; Baker, R.; Curtis, N. R.; Mawer, I. M.; Moseley, A. M.; Ridgill, M. P.; Rowley, M.; Stansfield, I.; Leeson, P. D., 3'-(Arylmethyl)- and 3'-(Aryloxy)-3phenyl-4-hydroxyquinolin- 2(1H)-ones: Orally Active Antagonists of the Glycine Site on the NMDA Receptor. Journal of Medicinal Chemistry 1994, 37 (10), 1402-1405. Quartaroli, M.; Carignani, C.; Dal Forno, G.; Mugnaini, M.; Ugolini, A.; Arban, R.; Bettelini, L.; Maraia, G.; Belardetti, F.; Reggiani, A.; Trist, D. G.; Ratti, E.; Di Fabio, R.; Corsi, M., Potent Antihyperalgesic Activity without Tolerance Produced by Glycine Site Antagonist of N-Methyl-D-Aspartate Receptor GV196771A. The Journal of Pharmacology and Experimental Therapeutics 1999, 290 (1), 158-169. Sun, L.; Tran, N.; Tang, F.; App, H.; Hirth, P.; McMahon, G.; Tang, C., Synthesis and Biological Evaluations of 3-Substituted Indolin-2-ones:  A Novel Class of Tyrosine Kinase Inhibitors That Exhibit Selectivity toward Particular Receptor Tyrosine Kinases. Journal of Medicinal Chemistry 1998, 41 (14), 2588-2603. Monge, A.; Palop, J.; Ramirez, C.; Font, M.; Fernandez-Alvarez, E., New 5H-1,2,4triazino[5,6-b]indole and aminoindole derivatives. Synthesis and studies as inhibitors of blood platelet aggregation, anti-hypertensive agents and thromboxane synthetase inhibitors. European Journal of Medicinal Chemistry 1991, 26 (2), 179-188. (a) Huang, J.; May, J. M., Ascorbic acid protects SH-SY5Y neuroblastoma cells from apoptosis and death induced by [beta]-amyloid. Brain Research 2006, 1097 (1), 52-58; (b) Lai, C.-T.; Yu, P. H., Dopamine- and -[beta]-3,4-dihydroxyphenylalanine hydrochloride-(-Dopa)-induced cytotoxicity towards catecholaminergic neuroblastoma SH-SY5Y Cells : Effects of oxidative stress and antioxidative factors. Biochemical Pharmacology 1997, 53 (3), 363-372; (c) Somayajulu, M.; McCarthy, S.; Hung, M.; Sikorska, M.; Borowy-Borowski, H.; Pandey, S., Role of mitochondria in neuronal cell death induced by oxidative stress; neuroprotection by Coenzyme Q10. Neurobiology of Disease 2005, 18 (3), 618-627. Ng, T. B.; Liu, F.; Wang, Z. T., Antioxidative activity of natural products from plants. Life Sciences 2000, 66 (8), 709-723. Watson, G. B.; Bolanowski, M. A.; Baganoff, M. P.; Deppeler, C. L.; Lanthorn, T. H., d-Cycloserine acts as a partial agonist at the glycine modulatory site of the NMDA receptor expressed inXenopus oocytes. Brain Research 1990, 510 (1), 158-160. Hollmann, M.; Heinemann, S., Cloned Glutamate Receptors. Annual Review of Neuroscience 1994, 17 (1), 31-108. (a) Birnstiel, S.; Wülfert, E.; Beck, S. G., Levetiracetam (ucb LO59) affects in vitro models of epilepsy in CA3 pyramidal neurons without altering normal synaptic transmission. Naunyn-Schmiedeberg's Archives of Pharmacology 1997, 356 (5), 611618; (b) Mazarati, A. M.; Baldwin, R.; Klitgaard, H.; Matagne, A.; Wasterlain, C. G., Anticonvulsant effects of levetiracetam and levetiracetam-diazepam combinations in experimental status epilepticus. Epilepsy Research 2004, 58 (2-3), 167-174. Pyo, M. K.; Jin, J. L.; Koo, Y. K.; Yun-Choi, H. S., Phenolic and Furan type compounds Isolated From Gastrodia elata and their anti-platelet effects. Archives of Pharmacal Research 2004, 27 (4), 381-385. Noda, N.; Kobayashi, Y.; Miyahara, K.; Fukahori, S., 2,4-Bis(4-hydroxybenzyl) phenol from Gastrodia elata. Phytochemistry 1995, 39 (5), 1247-1248. Tang, W.; Hioki, H.; Harada, K.; Kubo, M.; Fukuyama, Y., Antioxidant Phenylpropanoid-Substituted Epicatechins from Trichilia catigua. Journal of Natural Products 2007, 70 (12), 2010-2013. Zheng, W.; Wang, S. Y., Antioxidant Activity and Phenolic Compounds in Selected Herbs. Journal of Agricultural and Food Chemistry 2001, 49 (11), 5165-5170. 178 126. Cai, Y.; Luo, Q.; Sun, M.; Corke, H., Antioxidant activity and phenolic compounds of 112 traditional Chinese medicinal plants associated with anticancer. Life Sciences 2004, 74 (17), 2157-2184. 127. (a) Low, C.-M.; Lyuboslavsky, P.; French, A.; Le, P.; Wyatte, K.; Thiel, W. H.; Marchan, E. M.; Igarashi, K.; Kashiwagi, K.; Gernert, K.; Williams, K.; Traynelis, S. F.; Zheng, F., Molecular Determinants of Proton-Sensitive N-Methyl-D-aspartate Receptor Gating. Molecular Pharmacology 2003, 63 (6), 1212-1222; (b) Low, C.-M.; Zheng, F.; Lyuboslavsky, P.; Traynelis, S. F., Molecular determinants of coordinated proton and zinc inhibition of N-methyl-d-aspartate NR1/NR2A receptors. Proceedings of the National Academy of Sciences of the United States of America 2000, 97 (20), 11062-11067; (c) Traynelis, S. F.; Burgess, M. F.; Zheng, F.; Lyuboslavsky, P.; Powers, J. L., Control of Voltage-Independent Zinc Inhibition of NMDA Receptors by the NR1 Subunit. The Journal of Neuroscience 1998, 18 (16), 6163-6175. 179 [...]... project, the endeavour to search for potential antiepileptic and neuroprotective agents will be made on natural products, exemplified by both Gou Teng and Tian Ma, and synthetic compounds tentatively targeting at NMDA receptor In the following sections, the current trend in drug discovery from natural products, previous phytochemical and pharmacological studies on Gou Teng and Tian Ma, NMDA antagonism as a. .. used for the treatment of epilepsy constitute both Gou Teng and Tian Ma Moreover, a recent investigation has clearly indicated that co-application of extracts derived from Gou Teng and Tian Ma would significantly delay the onset time of wet dog shakes (WDS) in kainic-acid treated rats3 Besides, polypharmacy is a widely employed strategy in the pharmacotherapy of epilepsy due to the complexity in classification... 3 Pre-fractionated natural products library prepared from plants 9 4 Methodology adopted to obtain the standardized extracts from Gou Teng and Tian Ma 43 5 Preparation of the alkaloidal extract 45 6 Preparation of the EFME from Tian Ma 63 7 Retro-synthesis of BHE, HFC and CSD 75 8 Retro-synthesis of HPMP 75 9 Synthesis of HPMP 76 10 Conversion of 4,4’-diaminophenyl methane to DHPM by diazotization/hydrolysis... chromatography on silica column Followed by polyamide cartridge Flash fractions 2-5 Flash fractions 6 Parallel four-channel prep-HPLC 40 sub-fractions for each flash fraction Parallel eight-channel HPLC-ELSD-MS analysis HTS Active sub-fractions Purification and characterization Novel pharmaceutical lead Scheme 3 Pre-fractionated natural products library prepared from plants 11 The organic and aqueous... physicochemical properties which are beneficial to group and analyze them when extraction and separation are carried out However, the drawbacks of this approach are apparent as well First, purification of one chemical class from natural products may not be suitable for the HTS Secondly, it may also ignore the possible interaction present in these structurally related structures Last but not the least, the assumption... FIGURE PAGE 1 Qualitative pharmacophore for antagonists at Gly/NMDAR based on the structure of CTHQ 36 2 A developed pharmacophore model 37 3 Substituted 2-oxindoles proposed as potential antagonistic effect at Gly/NMDAR 38 4 EMS scans for peak 1 in TIC of the alkaloidal extract 46 5 Total ion chromatogram (TIC) of the alkaloidal extract (A) ; UV spectrum of the alkaloidal extract (B) 47 6 General structure... site as a partial agonist 123 xiv LIST OF TABLES TABLE PAGE 1 in vitro & in vivo model for epileptogenesis 32 2 Estimated MW and proposed structures for each component in alkaloidal extract 47 3 13 54 4 Quantities of five major alkaloids in the crude extract and in the alkaloidal extract of Gou Teng 59 5 Proposed structures and their characteristic ion fragments 64 6 ED50 of the standardized extracts for. .. of alkaloids1 The constituents of Tian Ma which are mainly small phenolic compounds were studied since the 1980s 2 On the basis of these phytochemical investigations, other researchers have either employed crude extracts 1 or single major constituents to study the pharmacological actions of these two herbs The reported activity can vary greatly, from antiepileptic to antiproliferative; from vasodilating... agents, two most famous traditional Chinese medicines (TCMs) which are monographed in Chinese Pharmacopoeia with antiepileptic effects, Gou Teng (Latin name: Ramulus Uncariae Cum Uncis) and Tian Ma (Latin name: Rhizoma Gastrodiae) have drawn our attention Since 1970s, J.D Phillipson and his co-workers have started the phytochemical study on the genus of Uncaria, and their focus was mainly on defining interesting... chemistry that a great variety of structures with “druglikeness” can be generated with less effort than natural products has diverted the attention of large pharmaceutical companies; third, the decision-makers in the large pharmaceutical companies has placed less focus on research in the area of infectious diseases, which traditionally has been a strong area of discovering new antibiotics from natural products7 . Methodology adopted to obtain the standardized extracts from Gou Teng and Tian Ma. 43 5 Preparation of the alkaloidal extract 45 6 Preparation of the EFME from Tian Ma. 63 7 Retro-synthesis. new antiepileptic and neuroprotective agents. Our aim is to search for active small molecules or molecular combinations from Gou Teng and Tian Ma, and to design and synthesize new NMDA receptor. A COMBINATORIAL APPROACH TO THE SEARCH FOR ANTICONVULSANT AGENTS FROM GOU TENG AND TIAN MA YANG HONG (B. Sc. (Pharm.)), Sichuan Univ. (M. Sc. (Pharm.)), Sichuan Univ.

Ngày đăng: 11/09/2015, 21:27

Từ khóa liên quan

Mục lục

  • Title page.pdf

  • part 1_20100116

  • Part 2_20100127

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan