1. Trang chủ
  2. » Giáo Dục - Đào Tạo

Anti inflammatory pharmacology of histone deacetylase inhibitors in pre clinical models of rheumatoid arthritis

233 483 0

Đang tải... (xem toàn văn)

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

THÔNG TIN TÀI LIỆU

ANTI-INFLAMMATORY PHARMACOLOGY OF HISTONE DEACETYLASE INHIBITORS IN PRE-CLINICAL MODELS OF RHEUMATOID ARTHRITIS CHOO QIUYI (B.Sc.(Pharmacy)(Hons.), NUS) A THESIS SUBMITTED FOR THE DEGREE OF DOCTOR OF PHILOSOPHY DEPARTMENT OF PHARMACY NATIONAL UNIVERSITY OF SINGAPORE 2010 Dedication This thesis is dedicated to my parents for nurturing me, without whom I would not be who I am this day. In addition, I would like to thank my parents and siblings for their unconditional support. Every little step that I take, I know you will be there for me as I will be there for you, to rejoice together when I succeed and to pick me up when I fall. It is also dedicated to my significant other for his love. Your presence has given me the strength and confidence to things I never thought I could. Thank you for being there for me whenever I needed you, even when I added on to the competing demands of a budding career, study and personal development. Acknowledgements I am grateful to NUS for providing the Research Scholarship so that I can be financially independent. Experiments had been undertaken at NUS with the support from the Department of Pharmacy. In particular, my gratification extends to my mentors A/P Paul Ho Chi Lui and Dr Lin Haishu, for allowing the study to be executed in the laboratories, their guidance along the way and many stimulating discussions. Publications and Conference Abstracts Publications The following manuscripts arose based on this thesis: 1. Choo QY1, Ho PC1, Lin HS1. Histone Deacetylase Inhibitors: New Hope for Rheumatoid Arthritis. Current Pharmaceutical Design 2008;14:803-820. 2. Choo QY1, Ho PC1, Tanaka Y2, Lin HS1. Histone Deacetylase Inhibitors MS-275 and SAHA Induced Growth Arrest and Suppressed Lipopolysaccharide-Stimulated NF-κB p65 Nuclear Accumulation in Human Rheumatoid Arthritis Synovial Fibroblastic E11 Cells. Rheumatology 2010. DOI:10.1093/rheumatology/keq108 3. Choo QY1, Ho PC1, Tanaka Y2, Lin HS1. Histone Deacetylase Inhibitors MS-275 and SAHA Suppress p38 Mitogen Activated Protein Kinase Signaling Pathway and Chemotaxis in Rheumatoid Arthritic Synovial Fibroblastic E11 Cells. (Manuscript submitted for review). 4. Choo QY1, Ho PC1, Tanaka Y2, Lin HS1. Anti-inflammatory mechanisms of Belinostat: Suppression of Nuclear Factor-kappa B and Mitogen-Activated Protein Kinase Signaling Pathways. (Manuscript in preparation). 5. Choo QY1, Ho PC1, Lin HS1. Anti-inflammatory Mechanisms of Action of Histone Deacetylase Inhibitors in Auto-Immune Diseases: A Recent Update. (Manuscript in preparation). Contribution to publications for other research projects during candidature: 1. Lin HS1, Zhang W1, Go ML1, Choo QY1, Ho PC1. Determination of Z-3,5,4’trimethoxystilbene in rat plasma by a simple HPLC method: application in a pre-clinical pharmacokinetic study. Journal of Pharmaceutical and Biomedical Analysis. DOI: 10.1016/j.jpba.2010.03.028. 2. Lin HS1, Choo QY1, Ho PC1. Quantification of oxyresveratrol analog trans-2,4,3’,5’tetramethoxystilbene in rat plasma by a rapid HPLC method: application in a pre-clinical pharmacokinetic study. Biomedical Chromatography. DOI: 10.1002/bmc.1454 Conference Abstracts The following conference abstracts were presented based on this thesis: 1. Choo QY1, Yang J1, Ho PC1, Chan SY1, Lin HS1. In-Vitro Anti-Inflammatory Activities of PXD-101. 7th Biennial Globalization of Pharmaceutics Education Network (GPEN) Conference. Katholieke Universiteit, Utrecht University, Leiden University, Leuven, Belgium. 9th to 12th September, 2008. 2. Choo QY1, Yang J1, Ho PC1, Chan SY1, Lin HS1. Suppressive Effects of Histone Deacetylase Inhibitiors PXD-101 and TSA on Pro-Inflammatory Cytokines and Nitric Oxide Secretion. Chromatin Conference: Histones, Nucleosomes, Chromosomes and Genomes by Abcam Inc. Singapore. 9th February, 2009. 3. Choo QY1, Tanaka Y2, Ho PC1, Lin HS1. Anti-Inflammatory Mechanisms of Histone Deacetylase Inhibitor – Trichostatin A. PharmSci@Asia. China Pharmaceutical University, Nanjing, China. 27th May, 2009. 4. Choo QY1, Ho PC1, Tanaka Y2, Lin HS1. Histone Deacetylase Inhibitors SAHA and MS-275 Induced Growth Arrest, Suppressed NF-κB Activation, Down-Regulated the Secretions of Nitric Oxide, IL-6, IL-18, VEGF and MMPs in Rheumatoid Arthritis Synovial Fibroblast-Like Cells. 73rd Annual Scientific Meeting of the American College of Rheumatology by American College of Rheumatology. Pennsylvania, USA. 16th to 21st October, 2009. 5. Choo QY1, Ho PC1, Lin HS1. Anti-Inflammatory Mechanisms of Histone Deacetylase Inhibitor Trichostatin A: Suppression of NF-κB Activation. AAPS Annual Meeting and Exposition by American Association of Pharmaceutical Scientists. Los Angeles, USA. 8th to 12th November, 2009. 6. Choo QY1, Ho PC1, Tanaka Y2, Lin HS1. Anti-Inflammatory Activities of Histone Deacetylase Inhibitors MS-275 and SAHA: Suppression of NF-κB Activation. BioPharma Asia Convention 2010 by Terrapin. Singapore. 17th to 18th March, 2010. Department of Pharmacy, National University of Singapore, Singapore and 2First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan. Table of Contents Chapter Page 1. INTRODUCTION 1.1 Rheumatoid Arthritis 1.2 The Histone Code and HDAC Inhibitors 1.3 Pharmacological Activities of HDAC Inhibitors in Inflammation 14 1.4 Anti-Rheumatic Activities of HDAC Inhibitors in Pre-Clinical RA Models 18 2. HYPOTHESES AND OBJECTIVES 47 3. EFFECTS OF HDAC INHIBITORS ON RASF PROLIFERATION 50 3.1 Materials and Methods 3.1.1 Materials 50 50 A. Cell Line and Cell Culture 50 B. HDAC Inhibitors and Methotrexate (MTX) 50 C. Cell Proliferation Assay 50 D. Cytoxicity Assay 50 E. Flow Cytometry Analysis 51 F. Western Blot Analysis 51 3.1.2 Methods 52 A. Cell Proliferation Assay 52 B. Cytoxicity Assay 52 C. Interaction between HDAC Inhibitors and MTX 53 D. Flow Cytometry Analysis 54 E. Western Blot Analysis for p16, p21 and p27 Proteins 55 F. Calculations 56 3.2 Results 58 A. HDAC Inhibitors Induced Growth Arrest of E11 RASF-like Cells in a Concentration-Dependent but Non-Cytotoxic Manner 57 B. Combination of HDAC Inhibitors with MTX can be Synergistic for E11 Growth Arrest 61 C. HDAC Inhibitors cause E11 Gowth Arrest by Inducing Cell Cycle Arrest D. HDAC Inhibitors Up-Regulated p21Expression 3.3 Discussions 63 66 67 4. GENE PROFILING AFTER TREATMENT WITH HDAC INHIBITORS 4.1 Materials and Methods 4.1.1 Materials 70 70 70 A. Cell Line and Cell Culture 70 B. RNA Extraction 70 4.1.2 Methods 71 A. RNA Extraction and Quality Assessment 71 B. Microarray Hybridization and Image Acquisition 72 C. Microarray Data Mining 73 D. Pathway Analysis for Differentially Expressed Genes in E11 Cells 4.2 Results 73 75 A. HDAC Inhibitors Induced Gene Expression Changes in E11 RASF-like Cells 75 B. Network and Pathway Analysis of Differentially Expressed Genes 83 4.3 Discussions 88 5. EFFECTS OF HDAC INHIBITORS ON NF-κB AND MAPK SIGNALING PATHWAYS 91 5.1 Materials and Methods 5.1.1 Materials 91 91 A. Cell Lines and Cell Culture 91 B. NF-κB Assay 91 C. Co-IP Analysis 91 D. Western Blot Analysis 91 5.1.2 Methods A. NF-κB Assay 93 93 B. Co-IP Analysis for the Association between NF-κB p65 and p300 as well as yje Association between MKP-1 and p38α 93 C. Western Blot Analysis for the Distribution of Acetylated NF-κB p65 as well as for MKP-1, p38α and p-p38 expression D. Calculations 5.2 Results A. HDAC Inhibitors Inhibited LPS-induced NF-κB p65 94 94 95 Nuclear Accumulation in E11 RASF-like and THP-1 Monocyte-like Cells 95 B. HDAC Inhibitors Increased the Association between NF-κB p65 and p300 98 C. HDAC Inhibitor Increased Acetylated NF-κB p65 Accumulation in the Cytoplasm 99 D. HDAC Inhibitor-Treated Cells Expressed more MKP-1 and less p38α 101 E. HDAC Inhibitors Increased the Association between MKP-1 and p38α 5.3 Discussions 103 104 6. EFFECTS OF HDAC INHIBITORS ON PRO-INFLAMMATORY CYTOKINE AND NO SECRETION 6.1 Materials and Methods 6.1.1 Materials 111 111 111 A. Cell Lines and Cell Culture 111 B. Hydrocortisone (HYD) and BAY 11-7082 111 C. ELISA Sets 111 D. NO Assay 111 E. Reverse Transcriptase-Polymerase Chain Reaction (RT-PCR) 6.1.2 Methods 111 113 A. Cytokine Secretion 113 B. NO Assay 113 C. Interaction between HDAC Inhibitors and MTX 114 D. RT-PCR analysis for COX-2 and iNOS Expression 115 E. Calculations 116 6.2 Results 117 A. HDAC Inhibitors Suppressed Pro-Inflammatory Cytokines in E11 RASF-like and THP-1 Monocyte-like Cells in a Concentration-Dependent Manner 117 B. HDAC Inhibitors Suppressed NO in E11 RASF-like and RAW264.7 Macrophage-like Cells in a ConcentrationDependent Manner 127 C. Combination of HDAC Inhibitor with MTX can be Synergistic for NO Suppression 131 D. HDAC Inhibitors Reduced COX-2 and iNOS Expression in E11 Cells 6.3 Discussions 134 135 7. EFFECTS OF HDAC INHIBITORS ON OTHER DOWNSTREAM EFFECTORS OF NF-κB AND MAPK SIGNALING PATHWAYS 7.1 Materials and Methods 7.1.1 Materials 140 140 140 A. Cell Lines and Cell Culture 140 B. VEGF Assay 140 C. HUVEC Angiogenesis Assay 140 D. Chemotaxis under Agarose 140 E. Chemokine ELISA 141 F. Substrate Gel Zymography 7.1.2 Methods 141 142 A. VEGF Assay 142 B. HUVEC Angiogenesis Assay 142 C. Chemotaxis ELISA 143 D. Chemotaxis under Agarose 143 E. Substrate Gel Zymography for MMP-2 and MMP-9 in E11 cells 7.2 Results 144 146 A. HDAC Inhibitors Down-Regulated VEGF in E11 RASF-like Cells in a concentration-dependent manner 147 B. HDAC Inhibitors Totally Abrogated VEGF-induced HUVEC Angiogenesis 148 C. HDAC Inhibitors Suppressed Chemokines in E11 Monocytelike Cells in a Concentration-Dependent Manner D. HDAC Inhibitors Reduced Monocyte Chemotaxis 149 154 E. HDAC Inhibitors Down-Regulated MMP-2 and MMP-9 Expression in E11 RASF-like Cells 7.3 Discussions 8. CONCLUSIONS AND FUTURE PERSPECTIVES 156 157 160 8.1 Conclusions 160 8.2 Future Perspectives 162 8.2.1 Determine the Relationship between Epigenetics and RA 162 144. Kiernan R, Bres V, Ng RWM, Coudart MP, El Messaoudi S, Sardet C, et al. Post- activation turn-off of NF-kappa B-dependent transcription is regulated by acetylation of p65. level. Biochemical and Biophysical Research Communications. 1997; 230(1):44-48. 145. Portanova P, Russo T, Pellerito O. The role of oxidative stress in apoptosis induced by the histone deacetylase inhibitor suberoylanilide hydroxamic acid in human colon adenocarcinoma HT-29 cells. International Journal of Oncology 2008; 33:325-31. 146. Feng R, Oton A, Mapara MY. The histone deacetylase inhibitor, PXD101, potentiates bortezomib-induced anti-multiple myeloma effect by induction of oxidative stress and DNA damage. Br J Haematology 2007; 139:385-97. 147. Schwab M, Reynders V, Ulrich S. PPARgamma is a key target of butyrate- induced caspase-3 activation in the colorectal cancer cell line Caco-2. Apoptosis 2006; 11:1801-11. 148. Hammaker D, Firestein GS. "Go upstream, young man'': lessons learned from the p38 saga. Annals of the Rheumatic Diseases. 2010 Jan; 69:77-82. 149. Yoshizawa T, Hammaker D, Boyle DL, Corr M, Flavell R, Davis R, et al. Role of MAPK Kinase in Arthritis: Distinct Mechanism of Action in Inflammation and Cytokine Expression. Journal of Immunology. 2009 ; 183(2):1360-1367. 150. Shaik SS, Soltau TD, Chaturvedi G, Totapally B, Hagood JS, Andrews WW, et al. Low Intensity Shear Stress Increases Endothelial ELR+ CXC Chemokine Production via a Focal Adhesion Kinase-p38 beta MAPK-NF-kappa B Pathway. Journal of Biological Chemistry. 2009; 284(9):5945-5955. 196 151. Guo X, Salmon RA, Schrader JW. A novel strategy demonstrates conclusively the involvement of p38 alpha MAP kinase in the production of the anti-inflammatory cytokine IL-10. Faseb Journal. 2000; 14(6):A1083-A1083. 152. Zhao TC, Cheng G, Zhang LX. Inhibition of histone deacetylases triggers pharmacologic preconditioning effects against myocardial ischemic injury. Cardiovasclar Research 2007; 76:473-81. 153. Heo H, Yoo L, Shin KS. Suppression of caspase-11 expression by histone deacetylase inhibitors. Biochemical and Biophysical Research Communications 2009; 378:79-83. 154. Wang C, Deng L, Hong M, Akkaraju GR, Inoue J, Chen ZJJ. TAK1 is a ubiquitin-dependent kinase of MKK and IKK. Nature. 2001; 412(6844):346-351. 155. Fukushima A, Boyle DE, Corr M, Firestein GS. Kinetic analysis of synovial signaling and gene expression in animal models for rheumatoid arthritis. Annals of the Rheumatic Diseases. Published online first: 26 May 2009. doi: 10.1136/ard.2009.112201 156. Pierce JW, Schoenleber R, Jesmok G, Best J, Moore SA, Collins T, et al. Novel inhibitors of cytokine-induced I kappa B alpha phosphorylation and endothelial cell adhesion molecule expression show anti-inflammatory effects in vivo. Journal of Biological Chemistry. 1997; 272(34):21096-21103. 157. Cuenda A, Rouse J, Doza YN, Meier R, Cohen P, Gallagher TF, et al. SB-203580 is a specific inhibitor of a MAP kinase homolog which is stimulated by cellular stresses and interleukin-1. Febs Letters. 1995 May; 364(2):229-231. 197 158. Inche AG, La Thangue NB. Keynote review: Chromation control and cancer-drug discovery: realizing the promise. Drug Discovery Today. 2006; 11(3-4):97-109. 159. Liu T, Kuljaca S, Tee A, Marshall GM. Histone deacetylase inhibitors: multifunctional anticancer agents. Cancer treatment reviews. 2006; 32(3):157-165. 160. McInnes IB, Schett G. Cytokines in the pathogenesis of rheumatoid arthritis. Nature Reviews Immunology. 2007; 7:429-442. 161. van Vollenhoven RF, Emery P, Bingham C, Keystone E, Greenwalds M, Moreland LW, et al. Safety of rituximab in rheumatoid arthritis: Results of a pooled analysis. Annals of the Rheumatic Diseases. 2006; 65:332-332. 162. Genovese M, Breedveld F, Emery P, Cohen S, Keystone E, Matteson E, et al. Safety of other biologic therapies following rituximab treatment in RA patients. Arthritis and Rheumatism. 2008; 58(9):S785-S785. 163. Singh V, Mishra R, Pritchard CH. Is It Safe to Use Biologics after Rituximab Therapy? Arthritis and Rheumatism. 2008; 58(12):4017-4018. 164. Marra CA, Guh G, Fisher JH, Chalmers A, Esdaile JM, Anis AH. The effectiveness of cyclosporine (CyA) in rheumatoid arthritis (RA): A longitudinal analysis of a population-based registry. Arthritis and Rheumatism. 2000; 43(9):402. 165. Schnitzer TJ, Yocum DE, Michalska M, Balius R, Snider ME, Hays A, et al. Subcutaneous administration of CAMPATH(R)-1H: Clinical and biological outcomes. Journal of Rheumatology. 1997; 24(6):1031-1036. 166. Minucci S, Pelicci PG. Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nature Reviews Cancer. 2006; 6(1):38-51. 198 167. Keane J, Bresnihan B. Tuberculosis reactivation during immunosuppressive therapy in rheumatic diseases: diagnostic and therapeutic strategies. Current Opinion in Rheumatology. 2008; 20(4):443-449. 168. Gomez-Reino JJ, Carmona L, Descalzo MA, Grp B. Risk of tuberculosis in patients treated with tumor necrosis factor antagonists due to incomplete prevention of reactivation of latent infection. Arthritis & Rheumatism-Arthritis Care & Research. 2007; 57(5):756-761. 169. Takahashi Y, Sugiyama H, Yamashita H, Kunimatsu J, Shimizu A, Toshiki E, et al. A risk assessment of nontuberculous mycobacteriosis during tnf blocking therapies for rheumatoid arthritis. Arthritis and Rheumatism. 2008; 58(9):S557-S558. 170. Balkwill F. Tumor necrosis factor or tumor promoting factor? Cytokine & Growth Factor Reviews. 2002; 13(2):135-141. 171. http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/2008/ucm1169 42.htm - last accessed on 16 June 2010. 172. http://www.arthritistoday.org/news/cancer-warning-tnf-blockers-kids.php - last accessed on 16 June 2010 173. Theophile H, Kahn V, Lacoin C, Baronnet L, Schaeverbeke T, Haramburu F, et al. Anti-TNF exposure duration and lymphoma risk. Fundamental & Clinical Pharmacology. 2008 Jun; 22:367-375. 199 174. Nakahara H, Song H, Sugimoto M, Hagihara K, Kishimoto T, Yoshizaki K, et al. Anti-interleukin-6 receptor antibody therapy reduces vascular endothelial growth factor production in rheumatoid arthritis. Arthritis and Rheumatism. 2003; 48(6):1521-1529. 175. Nelson RD, Quie PG, Simmons RL. Chemotaxis under agarose - New and simple method for measuring chemotaxis and spontaneous migration of human polymorphonuclear leukocytes and monocytes. Journal of Immunology. 1975; 115(6):1650-1656. 176. Neumann E, Gay RE, Gay S, Muller-Ladner U. Functional genomics of fibroblasts. Current Opinion in Rheumatology. 2004; 16(3):238-245. 177. Pierer M, Rethage J, Seibl R, Lauener R, Brentano F, Wagner U, et al. Chemokine secretion of rheumatoid arthritis synovial Fibro blasts stimulated by toll-like receptor ligands. Journal of Immunology. 2004; 172(4):2704-2704. 178. Heussen C, Dowdle EB. Electrophoretic analysis of plasminogen activators in polyacrylamide gels contianing sodium dodecyl-dulfate and copolymerized substrates. Analytical Biochemistry. 1980; 102(1):196-202. 177. Takeda K, Akira S. TLR signaling pathways. Seminars in Immunology. 2004; 16(1):3-9. 178. Egeblad M, Werb Z. New functions for the matrix metalloproteinases in cancer progression. Nature Reviews Cancer. 2002; 2(3):161-174. 179. Jackson C, Nguyen M, Arkell J, Sambrook P. Selective matrix metalloproteinase (MMP) inhibition in rheumatoid arthritis - Targetting gelatinase A activation. Inflammation Research. 2001; 50(4):183-186. 200 180. Huber LC, Brock M, Hemmatazad H, Giger OT, Moritz F, Trenkmann M, et al. Histone deacetylase/acetylase activity in total synovial tissue derived from rheumatoid arthritis and osteoarthritis patients. Arthritis and Rheumatism. 2007; 56(4):1087-1093. 181. Ishikawa F, Miyoshi H, Nose K, Shibanuma M. Transcriptional induction of MMP-10 by TGF-β, mediated by activation of MEF2A and downregulation of class IIa HDACs. Oncogene. 2010;29:909-919. 182. Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science. 2003; 299(5609):1057-1061. 183. Fisson S, Darrasse-Jeze G, Litvinova E, Septier F, Klatzmann D, Liblau R, et al. Continuous activation of autoreactive CD4(+) CD25(+) regulatory T cells in the steady state. Journal of Experimental Medicine. 2003; 198(5):737-746. 184. Wang LQ, de Zoeten EF, Greene MI, Hancock WW. Immunomodulatory effects of deacetylase inhibitors: therapeutic targeting of FOXP3(+) regulatory T cells. Nature Reviews Drug Discovery. 2009; 8(12):969-981. 185. Wildin RS, Ramsdell F, Peake J, Faravelli F, Casanova JL, Buist N, et al. Mutations in the novel forkhead/winged-helix protein Scurfin cause neonatal diabetes, enteropathy, thrombocytopenia, and endocrinopathy syndrome, the human equivalent of the scurfy mouse. American Journal of Human Genetics. 2000; 67(4):151. 186. Ochs HD, Khattri R, Bennett CL, Brunkow ME. Immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome and the scurfy mutant mouse. Immunology and Allergy Clinics of North America. 2002; 22(2):357-+. 201 187. Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4(+)CD25(+) regulatory T cells. Nature Immunology. 2003; 4(4):330-336. 188. Riley JL, June CH, Blazar BR. Human T Regulatory Cell Therapy: Take a Billion or So and Call Me in the Morning. Immunity. 2009; 30(5):656-665. 189. Li B, Greene MI. FOXP3 actively represses transcription by recruiting the HAT/HDAC complex. Cell Cycle. 2007; 6(12):1432-1436. 190. Tao R, de Zoeten EF, Ozkaynak E, Chen CX, Wang LQ, Porrett PM, et al. Deacetylase inhibition promotes the generation and function of regulatory T cells. Nature Medicine. 2007; 13(11):1299-1307. 191. Reilly CM, Thomas M, Gogal R, Olgun S, Santo A, Sodhi R, et al. The histone deacetylase inhibitor trichostatin A upregulates regulatory T cells and modulates autoimmunity in NZB/W F1 mice. Journal of Autoimmunity. 2008; 31(2):123-130 192. Lucas JL, Mirshahpanah P, Haas-Stapleton E, Asadullah K, Zollner TM, Numerof RP. Induction of Foxp3(+) regulatory T cells with histone deacetylase inhibitors. Cellular Immunology. 2009; 257(1-2):97-104. 193. Wang LQ, Tao R, Hancock WW. Using histone deacetylase inhibitors to enhance Foxp3(+) regulatory T-cell function and induce allograft tolerance. Immunology and Cell Biology. 2009; 87(3):195-202. 194. de Zoeten EF, Wang LQ, Sai H, Dillmann WH, Hancock WW. Inhibition of HDAC9 Increases T Regulatory Cell Function and Prevents Colitis in Mice. Gastroenterology. 2010; 138(2):583-594. 202 195. Mai A, Massa S, Pezzi R, Simeoni S, Rotili D, Nebbioso A, et al. Class II (IIa)- selective histone deacetylase inhibitors. 1. Synthesis and biological evaluation of novel (aryloxopropenyl)pyrrolyl hydroxyamides. Journal of Medicinal Chemistry. 2005; 48(9):3344-3353. 196. Mai A, Massa S, Pezzi R, Rotili D, Loidl P, Brosch G. Discovery of (aryloxopropenyl)pyrrolyl hydroxyamides as selective inhibitors of class IIa histone deacetylase homologue HD1-A. Journal of Medicinal Chemistry. 2003; 46(23):48264829. 197. Jones P, Bottomley MJ, Carfi A, Cecchetti O, Ferrigno F, Lo Surdo P, et al. 2- trifluoroacetylthiophenes, a novel series of potent and selective class II histone deacetylase inhibitors. Bioorganic & Medicinal Chemistry Letters. 2008; 18(11):34563461. 198. Estiu G, Greenberg E, Harrison CB, Kwiatkowski NP, Mazitschek R, Bradner JE, et al. Structural origin of selectivity in class II-selective histone deacetylase inhibitors. Journal of Medicinal Chemistry. 2008; 51(10):2898-2906. 199. Mai A, Valente S, Nebbioso A, Simeoni S, Ragno R, Massa S, et al. New pyrrole- based histone deacetylase inhibitors: Binding mode, enzyme- and cell-based investigations. International Journal of Biochemistry & Cell Biology. 2009; 41(1):235247. 200. Haggarty SJ, Koeller KM, Wong JC, Grozinger CM, Schreiber SL. Domain- selective small-molecule inhibitor of histone deacetylase (HDAC6)-mediated tubulin 203 deacetylation. Proceedings of the National Academy of Sciences of the United States of America. 2003; 100(8):4389-4394. 201. Hancock WW, Wang LQ, de Zoeten EF, Bradner JE, Mazitschek R. HDAC6 is a key new epigenetic target for the enhancement of Treg production and function in vitro and in vivo. American Journal of Transplantation. 2008; 8:223-223. 202. Bardel E, Larousserie F, Charlot-Rabiega P, Coulomb-L'Hermine A, Devergne O. Human CD4(+)CD25(+)Foxp3(+) Regulatory T Cells Do Not Constitutively Express IL35. Journal of Immunology. 2008; 181(10):6898-6905. 203. Shevach EM. Mechanisms of Foxp3(+) T Regulatory Cell-Mediated Suppression. Immunity. 2009; 30(5):636-645. 204. Lawson JM, Tremble J, Dayan C, Beyan H, Leslie RDG, Peakman M, et al. Increased resistance to CD4(+)CD25(hi) regulatory T cell-mediated suppression in patients with type diabetes. Clinical and Experimental Immunology. 2008; 154(3):353359. 205. D'Alise AM, Auyeung V, Feuerer M, Nishio J, Fontenot J, Benoist C, et al. The defect in T-cell regulation in NOD mice is an effect on the T-cell effectors. Proceedings of the National Academy of Sciences of the United States of America. 2008; 105(50):19857-19862. 206. Horiuchi M, Morinobu A, Chin T, Sakai Y, Kurosaka M, Kumagai S. Expression and Function of Histone Deacetylases in Rheumatoid Arthritis Synovial Fibroblasts. Journal of Rheumatology. 2009; 36(8):1580-1589. 204 207. Egger G, Liang GN, Aparicio A, Jones PA. Epigenetics in human disease and prospects for epigenetic therapy. Nature. 2004; 429(6990):457-463. 208. Shiozawa K, Nakanishi T, Tan M, Fang HB, Wang WC, Edelman MJ, et al. Preclinical Studies of Vorinostat (Suberoylanilide Hydroxamic Acid) Combined with Cytosine Arabinoside and Etoposide for Treatment of Acute Leukemias. Clinical Cancer Research. 2009; 15(5):1698-1707. 209. O'Connor OA. Clinical experience with the novel histone deacetylase inhibitor vorinostat (suberoylanilide hydroxamic acid) in patients with relapsed lymphoma. British Journal of Cancer. 2006; 95:S7-S12. 210. Davis R, Peters DH, McTavish D. Valproic acid - A reappraisal of it pharmacological properties and clinical efficacy in epilepsy. Drugs. 1994; 47(2):332-372. 211. Gugler R, Unruh GEV. Clinical pharmacokinetics of valproic acid. Clinical Pharmacokinetics. 1980; 5(1):67-83. 212. Johannessen SI, Tomson T. Pharmacokinetic variability of newer antiepileptic drugs - When is monitoring needed? Clinical Pharmacokinetics. 2006; 45(11):1061-1075. 213. Kelly WK, O'Connor OA, Krug LM, Chiao JH, Heaney M, Curley T, et al. Phase I study of an oral histone deacetylase inhibitor, suberoylanilide hydroxamic acid, in patients with advanced cancer. Journal of Clinical Oncology. 2005; 23(17):3923-3931. 214. Drummond DC, Noble CO, Kirpotin DB, Guo ZX, Scott GK, Benz CC. Clinical development of histone deacetylase inhibitors as anticancer agents. Annual Review of Pharmacology and Toxicology. 2005; 45:495-528. 205 215. Shah MH, Binkley P, Chan K, Xiao J, Arbogast D, Collamore M, et al. Cardiotoxicity of histone deacetylase inhibitor depsipeptide in patients with metastatic neuroendocrine tumors. Clinical Cancer Research. 2006; 12(13):3997-4003. 216. Stadler WM, Margolin K, Ferber S, McCulloch W, Thompson JA. A phase II study of depsipeptide in refractory metastatic renal cell cancer. Clinical Genitourinary Cancer. 2006; 5(1):57-60. 217. Fournel M, Trachy-Bourget MC, Yan PT, Kalita A, Bonfils C, Beaulieu C, et al. Sulfonamide anilides, a novel class of histone deacetylase inhibitors, are antiproliferative against human tumors. Cancer Research. 2002; 62(15):4325-4330. 218. Spiller SE, Ravanpay AC, Hahn AW, Olson JM. Suberoylanilide hydroxamic acid is effective in preclinical studies of medulloblastoma. Journal of Neuro-Oncology. 2006; 79(3):259-270. 219. Moradei OM, Mallais TC, Frechette S, Paquin I, Tessier PE, Leit SM, et al. Novel aminophenyl benzamide-type histone deacetylase inhibitors with enhanced potency and selectivity. Journal of Medicinal Chemistry. 2007; 50:5543-5546. 220. Hamblett CL, Methot JL, Mampreian DM, Sloman DL, Stanton MG, Kral AM, et al. The discovery of 6-amino nicotinamides as potent and selective histone deacetylase inhibitors. Bioorganic & Medicinal Chemistry Letters. 2007; 17(19):5300-5309. 221. Jones P, Altamura S, Chakravarty PK, Cecchetti O, De Francesco R, Gallinari P, et al. A series of novel, potent, and selective histone deacetylase . Bioorganic & Medicinal Chemistry Letters. 2006; 16(23):5948-5952. 206 222. Curtin M, Glaser K. Histone deacetylase inhibitors: The Abbott experience. Current Medicinal Chemistry. 2003; 10(22):2373-2392. 223. Faller DV, Hermine O, Small T, Suarez F, O'Reilly R, Boulad F, et al. Phase I/II trial of Arginine Butyrate and ganciclovir in Epstein-Barr virus-associated lymphoid malignancies. Journal of Clinical Oncology. 2006; 24(18):432S-432S. 224. Douillard JY, Bennouna J, Vavasseur F, Deporte-Fety R, Thomare P, Giacalone F, et al. Phase I trial of interleukin-2 and high-dose arginine butyrate in metastatic colorectal cancer. Cancer Immunology Immunotherapy. 2000; 49(1):56-61. 225. Patnaik A, Rowinsky EK, Villalona MA, Hammond LA, Britten CD, Siu LL, et al. A phase I study of pivaloyloxymethyl butyrate, a prodrug of the differentiating agent butyric acid, in patients with advanced solid malignancies. Clinical Cancer Research. 2002; 8(7):2142-2148. 226. ClinialTrials.gov. A service of the US National Institute of Health. www.clinicaltrials.gov. Last acessed on 16 June 2010. 238. Reid T, Valone F, Lipera W, Irwin D, Paroly W, Natale R, et al. Phase II trial of the histone deacetylase inhibitor pivaloyloxymethyl butyrate (Pivanex, AN-9) in advanced non-small cell lung cancer. Lung Cancer. 2004; 45(3):381-386. 227. Maier S, Reich E, Martin R, Bachem M, Altug V, Hautmann RE, et al. Tributyrin induces differentiation, growth arrest and apoptosis in androgen-sensitive and androgenresistant human prostate cancer cell lines. International Journal of Cancer. 2000; 88(2):245-251. 207 228. Lin JQ, Gilbert J, Rudek MA, Zwiebel JA, Gore S, Jiemjit A, et al. A Phase I Dose-Finding Study of 5-Azacytidine in Combination with Sodium Phenylbutyrate in Patients with Refractory Solid Tumors. Clinical Cancer Research. 2009; 15(19):62416249. 229. Gore SD, Weng LJ, Zhai S, Figg WD, Donehower RC, Dover GJ, et al. Impact of the putative differentiating agent sodium phenylbutyrate on myelodysplastic syndromes and acute myeloid leukemia. Clinical Cancer Research. 2001; 7(8):2330-2339. 230. Undevia SD, Kindler HL, Janisch L, Olson SC, Schilsky RL, Vogelzang NJ, et al. A phase I study of the oral combination of CI-994, a putative histone deacetylase inhibitor, and capecitabine. Annals of Oncology. 2004; 15(11):1705-1711. 231. Kummar S, Gutierrez M, Gardner ER, Donovan E, Hwang K, Chung EJ, et al. Phase I trial of MS-275, a histone deacetylase inhibitor, administered weekly in refractory solid tumors and lymphoid malignancies. Clinical Cancer Research. 2007; 13(18):5411-5417. 232. Conley BA, Egorin MJ, Tait N, Rosen DM, Sausville EA, Dover G, et al. Phase I study of the orally administered butyrate prodrug, tributyrin, in patients with solid tumors. Clinical Cancer Research. 1998; 4(3):629-634. 233. Camacho LH, Olson J, Tong WP, Young CW, Spriggs DR, Malkin MG. Phase I dose escalation clinical trial of phenylbutyrate sodium administered twice daily to patients with advanced solid tumors. Investigational New Drugs. 2007; 25(2):131-138. 208 234. Furumai R, Matsuyama A, Kobashi N, Lee KH, Nishiyama N, Nakajima I, et al. FK228 (depsipeptide) as a natural prodrug that inhibits class I histone deacetylases. Cancer Research. 2002; 62(17):4916-4921. 235. Rasheed WK, Johnstone RW, Prince HM. Histone deacetylase inhibitors in cancer therapy. Expert Opinion on Investigational Drugs. 2007; 16(5):659-678. 236. Kraker AJ, Mizzen CA, Hartl BG, Miin J, Allis CD, Merriman RL. Modulation of histone acetylation by 4-(acetylamino)-N-(2-amino-phenyl) benzamide in HCT-8 colon carcinoma. Molecular Cancer Therapeutics. 2003; 2(4):401-408. 237. Edelman MJ, Bauer K, Khanwani S, Tait N, Trepel J, Karp J, et al. Clinical and pharmacologic study of tributyrin: an oral butyrate prodrug. Cancer Chemotherapy and Pharmacology. 2003; 51(5):439-444. 238. Hu ED, Dul E, Sung CM, Chen ZX, Kirkpatrick R, Zhang GF, et al. Identification of novel isoform-selective inhibitors within class I histone deacetylases. Journal of Pharmacology and Experimental Therapeutics. 2003; 307(2):720-728. 239. Beckers T, Burkhardt C, Wieland H, Gimmnich P, Ciossek T, Maier T, et al. Distinct pharmacological properties of second generation HDAC inhibitors with the benzamide or hydroxamate head group. International Journal of Cancer. 2007; 121(5):1138-1148. 240. Liu L, Chen BA, Qin SK, Li SY, He XM, Qiu SM, et al. A novel histone deacetylase inhibitor Chidamide induces apoptosis of human colon cancer cells. Biochemical and Biophysical Research Communications. 2010; 392(2):190-195. 209 241. Nimmanapalli R, Fuino L, Bali P, Gasparetto M, Glozak M, Tao JG, et al. Histone deacetylase inhibitor LAQ824 both lowers expression and promotes proteasomal degradation of Bcr-Abl and induces apoptosis of imatinib mesylate-sensitive or refractory chronic myelogenous leukemia-blast crisis cells. Cancer Research. 2003; 63(16):5126-5135. 242. de Bono JS, Kristeleit R, Tolcher A, Fong P, Pacey S, Karavasilis V, et al. Phase I Pharmacokineitic and Pharmacodynamic Study of LAQ824, a Hydroxamate Histone Deacetylase Inhibitor with a Heat Shock Protein-90 Inhibitory Profile, in Patients with Advanced Solid Tumors. Clinical Cancer Research. 2008; 14(20):6663-6673. 243. Ottmann OG, Deangelo DJ, Stone RM, Pfeifer H, Lowenberg B, Atadja P, et al. A phase 1, pharmacokinetic (PK) and pharmacodynamic (PD) study of a novel histone deacetylase inhibitor LAQ824 in patients with hematologic malignancies. Journal of Clinical Oncology. 2004 Jul; 22(14):3024. 244. George P, Bali P, Annavarapu S, Scuto A, Fiskus W, Guo F, et al. Combination of the histone deacetylase inhibitor LBH589 and the hsp90 inhibitor 17-AAG is highly active against human CML-BC cells and AML cells with activating mutation of FLT-3. Blood. 2005; 105(4):1768-1776. 245. Butler LM, Webb Y, Agus DB, Higgins B, Tolentino TR, Kutko MC, et al. Inhibition of transformed cell growth and induction of cellular differentiation by pyroxamide, an inhibitor of histone deacetylase. Clinical Cancer Research. 2001; 7(4):962-970. 210 246. Plumb JA, Finn PW, Williams RJ, Bandara MJ, Romero MR, Watkins CJ, et al. Pharmacodynamic response and inhibition of growth of human tumor xenografts by the novel histone deacetylase inhibitor PXD101. Molecular Cancer Therapeutics. 2003; 2(8):721-728. 247. Kutko MC, Glick RD, Butler LM, Coffey DC, Rifkind RA, Marks PA, et al. Histone deacetylase inhibitors induce growth suppression and cell death in human rhabdomyosarcoma in vitro. Clinical Cancer Research. 2003; 9(15):5749-5755. 248. Tong WG, Wei Y, Stevenson W, Kuang SQ, Fang ZH, Zhang M, et al. Preclinical antileukemia activity of JNJ-26481585, a potent second-generation histone deacetylase inhibitor. Leukemia Research. 2010; 34(2):221-228. 249. Bhalla S, David K, Balasubramanian S, Prachand S, Mauro L, Sirisawad M, et al. PCI-24781, a novel histone deacetylase inhibitor (HDACI), induces caspase-dependent apoptosis in Hodgkin lymphoma (HL) and non-Hodgkin lymphoma (NHL) cell lines and is synergistic in combination with bortezomib. Annals of Oncology. 2008; 19:161-161. 250. http://www.curis.com/AACR2008CUDC101liver.pdf - last accessed on 16 June 2010. 251. http://www.4sc.de/en/news/pdf/Ecco_Presentation_17_09_09_eng.pdf - last accessed on 16 June 2010. 211 [...]... of Histone Deacetylase Inhibitors in Pre- clinical Models of Rheumatoid Arthritis HDAC inhibitors have emerged as a novel class of anti- cancer agents Their antirheumatic activities had been documented in various pre- clinical RA models However, their anti- rheumatic mechanisms of action are not well elucidated The work that was carried out for this thesis aimed to elucidate the anti- inflammatory and anti- rheumatic... values of HDAC inhibitors on NF-κB p65 nuclear accumulation inhibition in E11 cells 5.2 IC50 values of HDAC inhibitors on NF-κB p65 nuclear accumulation inhibition in THP-1 cells 6.1 97 IC50 values of HDAC inhibitors on pro -inflammatory cytokine inhibition in THP-1 cells 6.2 96 122 IC50 values of HDAC inhibitors on pro -inflammatory cytokine inhibition in E11 cells 125 6.3 IC50 values of HDAC inhibitors. .. NO inhibition in RAW264.7 cells 129 6.4 IC50 values of HDAC inhibitors on NO inhibition in E11 cells 130 6.5 Combination indices for the drug combinations in RAW264.7 cells 133 6.6 Combination indices for the drug combinations in E11 cells 134 6.7 Relative densitometry readings 135 7.1 IC50 values of HDAC inhibitors on VEGF inhibition in E11 cells 147 7.2 HDAC inhibitors totally abrogated VEGF-induced... effects of HDAC inhibitors 44 2.1 Possible anti- inflammatory and anti- rheumatic mechanisms of HDAC inhibitors 48 3.1 HDAC inhibitors and MTX inhibited E11 cell proliferation in a concentrationdependent manner 57 3.2 Effects of HDAC inhibitors and MTX on LDH release from E11 cells 59 3.3 Median-drug-effect plots for combinations of HDAC inhibitors and MTX 62 3.4 A representative DNA histogram and HDAC inhibitors. .. values of HDAC inhibitors on GCP-2 inhibition in THP-1 cells 151 7.4 IC50 values of HDAC inhibitors on MCP-2 inhibition in THP-1 cells 152 7.5 IC50 values of HDAC inhibitors on MIF inhibition in THP-1 cells 153 List of Figures Figure Page 1.1 The integrated immune response and pathogenesis of RA 2 1.2 The histone acetylation status controls gene expression 5 1.3 Effects of IL-1 in RA 18 1.4 Effects of. .. 1.5 HDAC inhibitors can inhibit cell proliferation by modulating CDK inhibitors 26 1.6 Involvement of the NF-κB signaling pathway in RA 30 1.7 HDAC2 as an important co-repressor molecule for glucocorticoid-mediated suppression of NF-κB-driven inflammatory gene expression 33 1.8 The MAPK signaling pathway 35 1.9 Inhibition of cartilage damage and bone destruction by HDAC inhibitors 41 1.10 Anti- angiogenic... combinations of HDAC inhibitors with MTX in RAW264.7 cells 6.9 130 SB203580 suppressed NO in RAW264.7 and E11 cells in a concentrationdependent manner 6.8 128 HDAC inhibitors and MTX suppressed NO in E11 cells in a concentrationdependent manner 6.7 126 HDAC inhibitors and MTX suppressed NO in RAW264.7 cells in a concentration-dependent manner 6.6 124 BAY 11-7082 suppressed IL-6 and IL-18 in E11 cells in. .. HDAC inhibitors as innovative anti- cancer modalities In October 2006, US Food and Drug Administration approved the first HDAC inhibitor – vorinostat (suberoylanilide hydroxamic acid or SAHA) It is indicated for cutaneous T-cell lymphoma (CTCL) [14-16] At least twenty other HDAC inhibitors had entered Phase I clinical trials (Table 1.2) Besides being anti- neoplastic, HDAC inhibitors are promising antiinflammatory... mechanisms of action of HDAC inhibitors Inhibition of RASF proliferation, suppression of proinflammatory cytokines, chemokines and NO as well as down-regulation of angiogenesis, chemotaxis and MMPs may provide beneficial effects in RA The aforementioned effects may be a result of CDK inhibitor p21 up-regulation as well as MAPK and NF-κB inhibition Hence, HDAC inhibitors appear to be an innovative strategy... 6.5 123 HDAC inhibitors suppressed IL-6 and IL-18 in E11 cells in a concentrationdependent manner 6.4 119 BAY 11-7082 suppressed IL-1β, IL-6, IL-18 and TNF-α in THP-1 cells in a concentration-dependent manner 6.3 103 133 Median-drug-effect plots for the combinations of HDAC inhibitors with MTX in E11 cells 134 6.10 HDAC inhibitors reduced COX-2 and iNOS expression in E11 cells 135 7.1 HDAC inhibitors . Summary Anti-inflammatory Pharmacology of Histone Deacetylase Inhibitors in Pre-clinical Models of Rheumatoid Arthritis HDAC inhibitors have emerged as a novel class of anti-cancer. ANTI-INFLAMMATORY PHARMACOLOGY OF HISTONE DEACETYLASE INHIBITORS IN PRE-CLINICAL MODELS OF RHEUMATOID ARTHRITIS CHOO QIUYI (B.Sc.(Pharmacy)(Hons.),. 6.1 IC 50 values of HDAC inhibitors on pro-inflammatory cytokine inhibition in THP-1 cells 122 6.2 IC 50 values of HDAC inhibitors on pro-inflammatory cytokine inhibition in E11 cells 125

Ngày đăng: 11/09/2015, 09:16

Xem thêm: Anti inflammatory pharmacology of histone deacetylase inhibitors in pre clinical models of rheumatoid arthritis

TỪ KHÓA LIÊN QUAN

Mục lục

    Thesis chapter 1 - Introduction page 1-46

    Thesis chapter 2 - Hypotheses and objectives page 47-49

    Thesis chapter 3 - Proliferation page 50-69

    Thesis chapter 4 - Microarray page 70-90

    Thesis chapter 5 - NFkB and MAPK signalling pathways page 91-110

    Thesis chapter 6 - Pro-inflammatory cytokines and NO page 111-139

    Thesis chapter 7 - Other downstream effectors page 140-159

    Thesis chapter 8 - Conclusions and future perspectives page 160-175

    Final_thesis_all_chapters_280311

    Thesis chapter 1 - Introduction page 1-46

TÀI LIỆU CÙNG NGƯỜI DÙNG

  • Đang cập nhật ...

TÀI LIỆU LIÊN QUAN