Hydrogen sulfide and neurogenic inflammation in a murine model of polymicrobial sepsis

243 448 0
Hydrogen sulfide and neurogenic inflammation in a murine model of polymicrobial sepsis

Đang tải... (xem toàn văn)

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

Thông tin tài liệu

HYDROGEN SULFIDE AND NEUROGENIC INFLAMMATION IN A MURINE MODEL OF POLYMICROBIAL SEPSIS ANG SEAH FANG (B.Sc. (Hons.), National University of Singapore) A THESIS SUBMITTED FOR THE DEGREE OF DOCTOR OF PHILOSOPHY NUS GRADUATE SCHOOL FOR INTEGRATIVE SCIENCES AND ENGINEERING NATIONAL UNIVERSITY OF SINGAPORE 2011 ACKNOWLEDGEMENTS It is a pleasure to thank the many people who made this dissertation possible. First and foremost, I would like to express my deepest gratitude to my supervisor, Professor Madhav Bhatia, for giving me confidence and support to begin my Ph.D studies. Professor Bhatia offered me so much advice, patiently supervising me, and always guiding me in the right direction. His passion for research, his stringent scientific attitude, and his perseverant spirits have been of great value to me. Special thanks are also given to my other supervisor, Associate Professor Paul A. MacAry. He is the one who accepted me as his student without any hesitation during the critical period of my Ph.D studies. He helped me immensely by giving me encouragement, guidance, supervison, and understanding throughout this work. It is not sufficient to express my gratitude with only a few words. I would also like to thank my co-supervisor, Associate Professor Shabbir M. Moochhala, for his engaging and proactive guidance. He has shared with me his invaluable insights on the animal model of polymicrobial sepsis. I appreciate all his contributions of time and ideas to make my Ph.D experience productive and stimulating. NUS Graduate School for Integrative Sciences and Engineering (NGS) has extended a great deal of support and ensured that I received a quality graduate education that will put me on the forefront of global competition. For that, I am truly thankful to NGS for i providing me with a generous scholarship as well as ample opportunities to shine and learn as a graduate student. My heartfelt appreciation also goes to Associate Professor Khanna Sanjay. I would like to thank him for his unselfish and unfailing support as my dissertation adviser. I am very grateful to Shoon Mei Leng, our laboratory officer, for her willingness to always go the extra mile to help me in my experiments and for excellent help in technical procedures. I would also like to thank the staff of Department of Pharmacology, especially Ting Wee Lee and Koh Yoke Moi, who have extended their warmest help to facilitate my Ph.D project. Special thanks also go to my fellow laboratory mates, Dr. Akhil Hegde, Dr. He Min, Dr. Jenab Nooruddinbhai Sidhapuriwala, Dr. Pratima Shrivastava, Dr. Raina Devi Ramnath, Dr. Ramasamy Tamizhselvi, Dr. Sun Jia, Dr. Zhang Huili, Dr. Zhang Jing, Dr. Muthu Kumaraswamy Shanmugam, Dr. Zhi Liang, Dr. Lee Jan Hau, Andy Yeo Yee, Koh Yung Hua, Sagiraju Sowmya, Yada Swathi, Yeo Ai Ling, and Abel Damien Ang for insightful discussions, moral support, and encouragement. Last but not least, I am greatly indebted to my family members for their support, love, and understanding so that I could come so far in life. ii TABLE OF CONTENTS ACKNOWLEDGEMENTS i TABLE OF CONTENTS iii SUMMARY ix LIST OF TABLES xii LIST OF FIGURES xiii LIST OF ABBREVIATIONS xvi PUBLICATIONS xx CHAPTER I INTRODUCTION 1.1 General overview 1.2 H2S 1.2.1 Chemical properties of H2S 1.2.2 H2S Toxicity 1.2.3 Biosynthesis of H2S 1.2.4 Metabolism of H2S 1.2.5 Biological roles of H2S 1.2.5.1 Roles of H2S in CNS 10 1.2.5.1.1 Physiological roles of H2S in CNS 10 1.2.5.1.2 H2S in CNS diseases 12 1.2.5.2 Roles of H2S in cardiovascular system 15 1.2.5.2.1 Physiological roles of H2S in cardiovascular system 15 1.2.5.2.2 H2S in cardiovascular diseases 18 1.2.5.3 Roles of H2S in gastrointestinal system 21 1.2.5.3.1 Physiological roles of H2S in gastrointestinal system 21 1.2.5.3.2 H2S in gastrointestinal diseases 25 1.2.5.4 Roles of H2S in endocrine system 26 iii 1.2.5.5 Roles of H2S in reproductive system 28 1.2.5.6 Roles of H2S in inflammation 28 1.2.5.6.1 Pro-inflammatory roles of H2S 31 1.2.5.6.2 Anti-inflammatory roles of H2S 42 1.2.5.6.3 H2S and neurogenic inflammation 45 1.3 Sepsis 55 1.3.1 Definition of sepsis 55 1.3.2 Epidemiology of sepsis 57 1.3.3 Pathophysiology of sepsis 58 1.3.4 Animal models of sepsis 61 1.3.4.1 Toxemia models 61 1.3.4.2 Bacterial infection models 62 1.3.4.3 Host-barrier disruption models 63 CHAPTER II RESEARCH RATIONALE AND OBJECTIVES 66 2.1 Question of interest 66 2.2 Approach 68 2.3 Objectives 69 CHAPTER III H2S PROMOTES TRPV1-MEDIATED NEUROGENIC INFLAMMATION IN POLYMICROBIAL SEPSIS 70 3.1 Introduction 70 3.2 Materials and methods 71 3.2.1 Induction of sepsis 71 3.2.2 Measurement of MPO activity 73 3.2.3 Measurement of plasma H2S 73 3.2.4 Assay of liver CSE activity 74 3.2.5 Histopathological examination 74 3.2.6 Survival studies 75 iv 3.2.7 Statistics 3.3 Results 75 75 3.3.1 Capsazepine attenuates systemic inflammation and multiple organ damage in sepsis 75 3.3.2 Capsazepine has no effect on endogenous generation of H2S in sepsis 77 3.3.3 Capsazepine protects against mortality in CLP-induced sepsis 77 3.3.4 Capsazepine reverses the pro-inflammatory effects of NaHS in sepsis 78 3.3.5 Capsazepine protects against NaHS-augmented mortality in CLPinduced sepsis 79 3.3.6 Capsazepine has no effect on blockade of endogenous generation of H2S in sepsis 79 3.3.7 Capsazepine has no effect on PAG-mediated attenuation of proinflammatory effects of H2S in sepsis 80 3.3.8 Capsazepine has no effect on PAG-mediated protection of mortality in CLP-induced sepsis 81 3.4 Discussion 81 CHAPTER IV H2S PROMOTES TRPV1-MEDIATED NEUROGENIC INFLAMMATION IN POLYMICROBIAL SEPSIS THROUGH ENHANCEMENT OF SP PRODUCTION 98 4.1 Introduction 98 4.2 Materials and methods 100 4.2.1 Induction of sepsis 100 4.2.2 Measurement of SP levels 100 4.2.3 Cytokines, chemokines, and adhesion molecules analysis 101 4.2.4 Reverse transcriptase-polymerase chain reaction analysis 101 4.2.5 Measurement of pulmonary edema 102 4.2.6 Alanine aminotransferase and aspartate aminotransferase assay 102 4.2.7 Statistics 102 4.3 Results 4.3.1 Capsazepine attenuates endogenous SP concentrations in both septic and septic mice administrated with NaHS 103 103 v 4.3.2 The attenuated SP concentration correlates with reduced production of pro-inflammatory molecules in both septic and septic mice administrated with NaHS 104 4.3.3 Capsazepine protects against MODS in both septic and septic mice administrated with NaHS 105 4.3.4 Capsazepine has no effect on PAG-mediated attenuation of SP levels in sepsis 106 4.3.5 Inhibition of H2S formation impaired pro-inflammatory molecules production after septic injury, but capsazepine has no effect on them 106 4.3.6 Beneficial effects of capsazepine and PAG are not additive in protection against MODS in sepsis 107 4.4 Discussion 107 CHAPTER V H2S PROMOTES TRPV1-MEDIATED NEUROGENIC INFLAMMATION IN POLYMICROBIAL SEPSIS BY ACTIVATING ERK1/2 AND NF-ΚB SIGNALING PATHWAYS 136 5.1 Introduction 136 5.2 Materials and methods 138 5.2.1 Induction of sepsis 138 5.2.2 Nuclear extraction and measurement of NF-κB activation 138 5.2.3 Western immunoblot 138 5.2.4 Statistics 139 5.3 Results 140 5.3.1 Effect of capsazepine on ERK1/2 activation in H2S-induced neurogenic inflammation in sepsis 140 5.3.2 Effect of capsazepine on IκBα phosphorylation and degradation levels in H2S-induced neurogenic inflammation in sepsis 140 5.3.3 Effect of capsazepine on NF-κB activation in H2S-induced neurogenic inflammation in sepsis 141 5.4 Discussion 142 vi CHAPTER VI H2S AUGMENTS COX-2 AND PROSTAGLANDIN E2 METABOLITE PRODUCTION IN SEPSIS-EVOKED ACUTE LUNG INJURY BY A TRPV1 CHANNEL-DEPENDENT MECHANISM 152 6.1 Introduction 152 6.2 Materials and methods 153 6.2.1 Induction of sepsis 153 6.2.2 Measurement of COX-2 activity 154 6.2.3 Measurement of PGE2 metabolite levels 154 6.2.4 Western immunoblot 154 6.2.5 Measurement of MPO activity 154 6.2.6 Histopathogical examination 155 6.2.7 Cytokines, chemokines, and adhesion molecules analysis 155 6.2.8 Measurement of pulmonary edema 155 6.2.9 Survival studies 155 6.2.10 Statistics 155 6.3 Results 155 6.3.1 H2S regulates COX-2 levels in septic lungs in a TRPV1 channeldependent manner 155 6.3.2 The H2S-augmented, TRPV1-dependent COX-2 response correlates with concurrent PGE2 metabolite production following septic injury 156 6.3.3 COX-2 inhibition prevents H2S from aggravating ALI in sepsis 157 6.3.4 Blockade of H2S-mediated activation of COX-2 impaired proinflammatory cytokines, chemokines and adhesion molecules production in sepsis-induced ALI 158 6.3.5 Inhibition of COX-2 attenuates H2S-augmented PGE2 metabolite production in septic lungs 159 6.3.6 Inhibition of COX-2 protects against H2S-augmented, CLP-induced lethality, but has no effect on PAG-mediated protection of mortality in sepsis 159 6.4 Discussion 160 vii CHAPTER VII GENERAL DISCUSSION AND CONCLUSION 180 7.1 Significance of findings 180 7.2 Limitations of the study 184 7.3 Conclusions and future perspectives 185 REFERENCES 189 viii SUMMARY Hydrogen sulfide (H2S), a malodorous gas with the characteristic odor of rotten eggs, has been recognized as an important endogenous gaseous signaling molecule of the cardiovascular, gastrointestinal, genitourinary, and nervous systems. Besides acting as a potent vasodilator and an atypical neuromodulator, H2S is increasingly being established as a novel mediator of inflammation. However, the part played by H2S in modulating neurogenic inflammatory response in sepsis is not known. Therefore, this study aimed to investigate the role of H2S in mediating neurogenic inflammation in a mouse model of polymicrobial sepsis induced by cecal ligation and puncture (CLP). Of major significance in the development of neurogenic inflammation is the transient receptor potential vanilloid type (TRPV1) receptor, a non-selective cation channel found predominantly in primary sensory neurons. In particular, the results of the present study indicate that H2S promotes TRPV1-mediated neurogenic inflammation in sepsis. It was found that capsazepine, a selective receptor antagonist of TRPV1, significantly attenuated systemic inflammation and multiple organ damage caused by CLP-induced sepsis under the pro-inflammatory impact of H2S. Capsazepine also delayed the onset of lethality and protected against sepsis-associated mortality. Administration of sodium hydrosulfide, an H2S donor, exacerbated but capsazepine reversed deleterious effects of sepsis. In the presence of DL-propargylglycine, an inhibitor of endogenous H2S synthesis, capsazepine caused no further changes to the DL-propargylglycine-mediated attenuation of systemic inflammation, multiple organ damage, and mortality in sepsis. Moreover, capsazepine had no effect on endogenous ix [154] Spiller F, Orrico MI, Nascimento DC, et al. Hydrogen sulfide improves neutrophil migration and survival in sepsis via K+ATP channel activation. Am J Respir Crit Care Med 2010; 182(3): 360-8. [155] Bruce AN. Vasodilatory axon reflexes. Q J Exp Physiol 1913; 6: 339-54 [156] Bruce AN. Uber die Beziehung der Sensiblen Nervenendigungen zum Enzundungsvorgang. Arch Exp Pathol Pharmakol 1910; 63: 424-33. [157] Jancso N, Jancso-Gabor A, Szolcsanyi J. Direct evidence for neurogenic inflammation and its prevention by denervation and by pretreatment with capsaicin. Br J Pharmacol Chemother 1967; 31(1): 138-51. [158] Richardson JD, Vasko MR. Cellular mechanisms of neurogenic inflammation. J Pharmacol Exp Ther 2002; 302(3): 839-45. [159] Matthay MA, Ware LB. Can nicotine treat sepsis? Nat Med 2004; 10(11): 11612. [160] Geppetti P, Holzer P. Neurogenic inflammation. USA: CRC Press 1996. [161] Szallasi A, Cortright DN, Blum CA, Eid SR. The vanilloid receptor TRPV1: 10 years from channel cloning to antagonist proof-of-concept. Nat Rev Drug Discov 2007; 6(5): 357-72. [162] Schumacher MA. Transient receptor potential channels in pain and inflammation: therapeutic opportunities. Pain Pract 2010; 10(3): 185-200. [163] Alawi K, Keeble J. The paradoxical role of the transient receptor potential vanilloid receptor in inflammation. Pharmacol Ther 2010; 125(2): 181-95. [164] Szallasi A, Blumberg PM. Vanilloid (Capsaicin) receptors and mechanisms. Pharmacol Rev 1999; 51(2): 159-212. [165] O'Connor TM, O'Connell J, O'Brien DI, Goode T, Bredin CP, Shanahan F. The role of substance P in inflammatory disease. J Cell Physiol 2004; 201(2): 167-80. 206 [166] Julius D, Basbaum AI. Molecular mechanisms of nociception. Nature 2001; 413(6852): 203-10. [167] Von Euler US, Gaddum JH. An unidentified depressor substance in certain tissue extracts. J Physiol 1931; 72(1): 74-87. [168] Satake H, Kawada T. Overview of the primary structure, tissue-distribution, and functions of tachykinins and their receptors. Curr Drug Targets 2006; 7(8): 963-74. [169] Bhatia M. Hydrogen sulfide and substance P in inflammation. Antioxid Redox Signal 2010; 12(10): 1191-202. [170] Zhang Y, Lu L, Furlonger C, Wu GE, Paige CJ. Hemokinin is a hematopoieticspecific tachykinin that regulates B lymphopoiesis. Nat Immunol 2000; 1(5): 392-7. [171] Page NM, Bell NJ, Gardiner SM, et al. Characterization of the endokinins: human tachykinins with cardiovascular activity. Proc Natl Acad Sci U S A 2003; 100(10): 6245-50. [172] Kurtz MM, Wang R, Clements MK, et al. Identification, localization and receptor characterization of novel mammalian substance P-like peptides. Gene 2002; 296(1-2): 205-12. [173] Brimijoin S, Lundberg JM, Brodin E, Hokfelt T, Nilsson G. Axonal transport of substance P in the vagus and sciatic nerves of the guinea pig. Brain Res 1980; 191(2): 443-57. [174] Harmar A, Schofield JG, Keen P. Cycloheximide-sensitive synthesis of substance P by isolated dorsal root ganglia. Nature 1980; 284(5753): 267-9. [175] Graham GJ, Stevens JM, Page NM, et al. Tachykinins regulate the function of platelets. Blood 2004; 104(4): 1058-65. [176] Bellucci F, Carini F, Catalani C, et al. Pharmacological profile of the novel mammalian tachykinin, hemokinin 1. Br J Pharmacol 2002; 135(1): 266-74. 207 [177] Di Maria GU, Bellofiore S, Geppetti P. Regulation of airway neurogenic inflammation by neutral endopeptidase. Eur Respir J 1998; 12(6): 1454-62. [178] Sternberg EM. Neural regulation of innate immunity: a coordinated nonspecific host response to pathogens. Nat Rev Immunol 2006; 6(4): 318-28. [179] Bhatia M, Zhi L, Zhang H, Ng SW, Moore PK. Role of substance P in hydrogen sulfide-induced pulmonary inflammation in mice. Am J Physiol Lung Cell Mol Physiol 2006; 291(5): L896-904. [180] Bhatia M, Sidhapuriwala JN, Ng SW, Tamizhselvi R, Moochhala SM. Proinflammatory effects of hydrogen sulphide on substance P in caerulein-induced acute pancreatitis. J Cell Mol Med 2008; 12(2): 580-90. [181] Zhang H, Hegde A, Ng SW, Adhikari S, Moochhala SM, Bhatia M. Hydrogen sulfide up-regulates substance P in polymicrobial sepsis-associated lung injury. J Immunol 2007; 179(6): 4153-60. [182] American College of Chest Physicians/Society of Critical Care Medicine Consensus Conference: definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. Crit Care Med 1992; 20(6): 864-74. [183] Angus DC, Linde-Zwirble WT, Lidicker J, Clermont G, Carcillo J, Pinsky MR. Epidemiology of severe sepsis in the United States: analysis of incidence, outcome, and associated costs of care. Crit Care Med 2001; 29(7): 1303-10. [184] Vincent JL, Sakr Y, Sprung CL, et al. Sepsis in European intensive care units: results of the SOAP study. Crit Care Med 2006; 34(2): 344-53. [185] Martin GS, Mannino DM, Eaton S, Moss M. The epidemiology of sepsis in the United States from 1979 through 2000. N Engl J Med 2003; 348(16): 1546-54. [186] Rittirsch D, Flierl MA, Ward PA. Harmful molecular mechanisms in sepsis. Nat Rev Immunol 2008; 8(10): 776-87. 208 [187] Liu SF, Malik AB. NF-κB activation as a pathological mechanism of septic shock and inflammation. Am J Physiol Lung Cell Mol Physiol 2006; 290(4): L622L45. [188] Russell JA. Management of sepsis. N Engl J Med 2006; 355(16): 1699-713. [189] Esmon CT, Fukudome K, Mather T, et al. Inflammation, sepsis, and coagulation. Haematologica 1999; 84(3): 254-9. [190] Bone RC, Grodzin CJ, Balk RA. Sepsis: a new hypothesis for pathogenesis of the disease process. Chest 1997; 112(1): 235-43. [191] Fink MP, Heard SO. Laboratory models of sepsis and septic shock. J Surg Res 1990; 49(2): 186-96. [192] Deitch EA. Animal models of sepsis and shock: a review and lessons learned. Shock 1998; 9(1): 1-11. [193] Riedemann NC, Guo RF, Ward PA. The enigma of sepsis. J Clin Invest 2003; 112(4): 460-7. [194] Buras JA, Holzmann B, Sitkovsky M. Animal models of sepsis: setting the stage. Nat Rev Drug Discov 2005; 4(10): 854-65. [195] Copeland S, Warren HS, Lowry SF, et al. Acute inflammatory response to endotoxin in mice and humans. Clin Diagn Lab Immunol 2005; 12(1): 60-7. [196] Doi K, Leelahavanichkul A, Yuen PS, Star RA. Animal models of sepsis and sepsis-induced kidney injury. J Clin Invest 2009; 119(10): 2868-78. [197] Cross AS, Opal SM, Sadoff JC, Gemski P. Choice of bacteria in animal models of sepsis. Infect Immun 1993; 61(7): 2741-7. [198] Remick DG, Newcomb DE, Bolgos GL, Call DR. Comparison of the mortality and inflammatory response of two models of sepsis: lipopolysaccharide vs. cecal ligation and puncture. Shock 2000; 13(2): 110-6. 209 [199] Zantl N, Uebe A, Neumann B, et al. Essential role of gamma interferon in survival of colon ascendens stent peritonitis, a novel murine model of abdominal sepsis. Infect Immun 1998; 66(5): 2300-9. [200] Maier S, Traeger T, Entleutner M, et al. Cecal ligation and puncture versus colon ascendens stent peritonitis: two distinct animal models for polymicrobial sepsis. Shock 2004; 21(6): 505-11. [201] Rittirsch D, Hoesel LM, Ward PA. The disconnect between animal models of sepsis and human sepsis. J Leukoc Biol 2007; 81(1): 137-43. [202] Deitch EA. Rodent models of intra-abdominal infection. Shock 2005; 24 (Suppl 1): 19-23. [203] Wichterman KA, Baue AE, Chaudry IH. Sepsis and septic shock--a review of laboratory models and a proposal. J Surg Res 1980; 29(2): 189-201. [204] Hotchkiss RS, Tinsley KW, Karl IE. Role of apoptotic cell death in sepsis. Scand J Infect Dis 2003; 35(9): 585-92. [205] Ayala A, Chaudry IH. Immune dysfunction in murine polymicrobial sepsis: mediators, macrophages, lymphocytes and apoptosis. Shock 1996; Suppl 1: S27-38. [206] Dellinger RP, Levy MM, Carlet JM, et al. Surviving Sepsis Campaign: international guidelines for management of severe sepsis and septic shock: 2008. Crit Care Med 2008; 36(1): 296-327. [207] Remick DG. Pathophysiology of sepsis. Am J Pathol 2007; 170(5): 1435-44. [208] Geppetti P, Materazzi S, Nicoletti P. The transient receptor potential vanilloid 1: role in airway inflammation and disease. Eur J Pharmacol 2006; 533(1-3): 207-14. [209] Barnes PJ. Asthma as an axon reflex. Lancet 1986; 1(8475): 242-5. [210] Joos GF, Pauwels RA. Tachykinin receptor antagonists: potential in airways diseases. Curr Opin Pharmacol 2001; 1(3): 235-41. 210 [211] Liddle RA. The role of Transient Receptor Potential Vanilloid (TRPV1) channels in pancreatitis. Biochim Biophys Acta 2007; 1772(8): 869-78. [212] Szabo A, Czirjak L, Sandor Z, et al. Investigation of sensory neurogenic components in a bleomycin-induced scleroderma model using transient receptor potential vanilloid receptor- and calcitonin gene-related peptide-knockout mice. Arthritis Rheum 2008; 58(1): 292-301. [213] Heyland DK, Hopman W, Coo H, Tranmer J, McColl MA. Long-term healthrelated quality of life in survivors of sepsis. Short Form 36: a valid and reliable measure of health-related quality of life. Crit Care Med 2000; 28(11): 3599-605. [214] Rittirsch D, Huber-Lang MS, Flierl MA, Ward PA. Immunodesign of experimental sepsis by cecal ligation and puncture. Nat Protoc 2009; 4(1): 31-6. [215] Beresford IJ, Sheldrick RL, Ball DI, et al. GR159897, a potent non-peptide antagonist at tachykinin NK2 receptors. Eur J Pharmacol 1995; 272(2-3): 241-8. [216] Dakhama A, Park JW, Taube C, et al. Alteration of airway neuropeptide expression and development of airway hyperresponsiveness following respiratory syncytial virus infection. Am J Physiol Lung Cell Mol Physiol 2005; 288(4): L761-70. [217] Lau HY, Wong FL, Bhatia M. A key role of neurokinin receptors in acute pancreatitis and associated lung injury. Biochem Biophys Res Commun 2005; 327(2): 509-15. [218] Medhurst AD, Hay DW, Parsons AA, Martin LD, Griswold DE. In vitro and in vivo characterization of NK3 receptors in the rabbit eye by use of selective nonpeptide NK3 receptor antagonists. Br J Pharmacol 1997; 122(3): 469-76. [219] Okajima K, Harada N, Uchiba M, Isobe H. Activation of capsaicin-sensitive sensory neurons by carvedilol, a nonselective beta-blocker, in spontaneous hypertensive rats. J Pharmacol Exp Ther 2004; 309(2): 684-91. 211 [220] Labarca C, Paigen K. A simple, rapid, and sensitive DNA assay procedure. Anal Biochem 1980; 102(2): 344-52. [221] Siegel LM. A direct microdetermination for sulfide. Anal Biochem 1965; 11: 126-32. [222] Brown KA, Brain SD, Pearson JD, Edgeworth JD, Lewis SM, Treacher DF. Neutrophils in development of multiple organ failure in sepsis. Lancet 2006; 368(9530): 157-69. [223] Moore PK, Bhatia M, Moochhala S. Hydrogen sulfide: from the smell of the past to the mediator of the future? Trends Pharmacol Sci 2003; 24(12): 609-11. [224] Washtien W, Abeles RH. Mechanism of inactivation of gamma-cystathionase by the acetylenic substrate analogue propargylglycine. Biochemistry 1977; 16(11): 2485-91. [225] Botha AJ, Moore FA, Moore EE, Sauaia A, Banerjee A, Peterson VM. Early neutrophil sequestration after injury: a pathogenic mechanism for multiple organ failure. J Trauma 1995; 39(3): 411-7. [226] Zhou MY, Lo SK, Bergenfeldt M, et al. In vivo expression of neutrophil inhibitory factor via gene transfer prevents lipopolysaccharide-induced lung neutrophil infiltration and injury by a β2 integrin-dependent mechanism. J Clin Invest 1998; 101(11): 2427-37. [227] Kyriakides C, Jasleen J, Wang Y, Moore FD, Jr., Ashley SW, Hechtman HB. Neutrophils, not complement, mediate the mortality of experimental hemorrhagic pancreatitis. Pancreas 2001; 22(1): 40-6. [228] Iwamura H, Sato M, Wakitani K. Comparative study of glucocorticoids, cyclosporine A, and JTE-607 [(-)-Ethyl-N[3,5-dichloro-2-hydroxy-4-[2-(4- 212 methylpiperazin-1-yl)ethoxy]benzoyl]- L-phenylalaninate dihydrochloride] in a mouse septic shock model. J Pharmacol Exp Ther 2004; 311(3): 1256-63. [229] Davis KA, Santaniello JM, He LK, et al. Burn injury and pulmonary sepsis: development of a clinically relevant model. J Trauma 2004; 56(2): 272-8. [230] Wang P, Chaudry IH. Mechanism of hepatocellular dysfunction during hyperdynamic sepsis. Am J Physiol 1996; 270(5 Pt 2): R927-38. [231] Mudd SH, Finkelstein JD, Irreverre F, Laster L. Transsulfuration in mammals. Microassays and tissue distributions of three enzymes of the pathway. J Biol Chem 1965; 240(11): 4382-92. [232] Puneet P, Hegde A, Ng SW, et al. Preprotachykinin-A gene products are key mediators of lung injury in polymicrobial sepsis. J Immunol 2006; 176(6): 3813-20. [233] Zuege D, Suzuki S, Berthiaume Y. Increase of lung sodium-potassium-ATPase activity during recovery from high-permeability pulmonary edema. Am J Physiol 1996; 271(6 Pt 1): L896-909. [234] Pittet JF, Wiener-Kronish JP, McElroy MC, Folkesson HG, Matthay MA. Stimulation of lung epithelial liquid clearance by endogenous release of catecholamines in septic shock in anesthetized rats. J Clin Invest 1994; 94(2): 663-71. [235] Pittet JF, Griffiths MJ, Geiser T, et al. TGF-β is a critical mediator of acute lung injury. J Clin Invest 2001; 107(12): 1537-44. [236] Kew MC. Serum aminotransferase concentration as evidence of hepatocellular damage. Lancet 2000; 355(9204): 591-2. [237] Ng SW, Zhang H, Hegde A, Bhatia M. Role of preprotachykinin-A gene products on multiple organ injury in LPS-induced endotoxemia. J Leukoc Biol 2008; 83(2): 288-95. 213 [238] Beer S, Weighardt H, Emmanuilidis K, et al. Systemic neuropeptide levels as predictive indicators for lethal outcome in patients with postoperative sepsis. Crit Care Med 2002; 30(8): 1794-8. [239] Elekes K, Helyes Z, Nemeth J, et al. Role of capsaicin-sensitive afferents and sensory neuropeptides in endotoxin-induced airway inflammation and consequent bronchial hyperreactivity in the mouse. Regul Pept 2007; 141(1-3): 44-54. [240] Veronesi B, Oortgiesen M. The TRPV1 receptor: target of toxicants and therapeutics. Toxicol Sci 2006; 89(1): 1-3. [241] Pinsky MR, Vincent JL, Deviere J, Alegre M, Kahn RJ, Dupont E. Serum cytokine levels in human septic shock. Relation to multiple-system organ failure and mortality. Chest 1993; 103(2): 565-75. [242] Krishna M, Narang H. The complexity of mitogen-activated protein kinases (MAPKs) made simple. Cell Mol Life Sci 2008; 65(22): 3525-44. [243] Chang L, Karin M. Mammalian MAP kinase signalling cascades. Nature 2001; 410(6824): 37-40. [244] Ronco MT, Manarin R, Frances D, Serra E, Revelli S, Carnovale C. Benznidazole treatment attenuates liver NF-κB activity and MAPK in a cecal ligation and puncture model of sepsis. Mol Immunol 2011; 48(6-7): 867-73. [245] O'Sullivan AW, Wang JH, Redmond HP. NF-κB and p38 MAPK inhibition improve survival in endotoxin shock and in a cecal ligation and puncture model of sepsis in combination with antibiotic therapy. J Surg Res 2009; 152(1): 46-53. [246] Shen L, Mo H, Cai L, et al. Losartan prevents sepsis-induced acute lung injury and decreases activation of nuclear factor-κB and mitogen-activated protein kinases. Shock 2009; 31(5): 500-6. 214 [247] Brown MA, Jones WK. NF-κB action in sepsis: the innate immune system and the heart. Front Biosci 2004; 9: 1201-17. [248] Baeuerle PA, Baichwal VR. NF-κB as a frequent target for immunosuppressive and anti-inflammatory molecules. Adv Immunol 1997; 65: 111-37. [249] Williams DL, Ha T, Li C, Kalbfleisch JH, Laffan JJ, Ferguson DA. Inhibiting early activation of tissue nuclear factor-κB and nuclear factor interleukin with (1->3)-beta-D-glucan increases long-term survival in polymicrobial sepsis. Surgery 1999; 126(1): 54-65. [250] Jiang B, Xu S, Hou X, Pimentel DR, Brecher P, Cohen RA. Temporal control of NF-κB activation by ERK differentially regulates interleukin-1β-induced gene expression. J Biol Chem 2004; 279(2): 1323-9. [251] Roux PP, Blenis J. ERK and p38 MAPK-activated protein kinases: a family of protein kinases with diverse biological functions. Microbiol Mol Biol Rev 2004; 68(2): 320-44. [252] Choi MS, Lee SH, Cho HS, et al. Inhibitory effect of obovatol on nitric oxide production and activation of NF-κB/MAP kinases in lipopolysaccharide-treated RAW 264.7 cells. Eur J Pharmacol 2007; 556(1-3): 181-9. [253] Dumitru CD, Ceci JD, Tsatsanis C, et al. TNF-α induction by LPS is regulated posttranscriptionally via a Tpl2/ERK-dependent pathway. Cell 2000; 103(7): 1071-83. [254] Ropert C, Closel M, Chaves AC, Gazzinelli RT. Inhibition of a p38/stressactivated protein kinase-2-dependent phosphatase restores function of IL-1 receptorassociate kinase-1 and reverses Toll-like receptor 2- and 4-dependent tolerance of macrophages. J Immunol 2003; 171(3): 1456-65. 215 [255] Khan MA, Kang J, Steiner TS. Enteroaggregative Escherichia coli flagellininduced interleukin-8 secretion requires Toll-like receptor 5-dependent p38 MAP kinase activation. Immunology 2004; 112(4): 651-60. [256] Yang G, Cao K, Wu L, Wang R. Cystathionine gamma-lyase overexpression inhibits cell proliferation via a H2S-dependent modulation of ERK1/2 phosphorylation and p21Cip/WAK-1. J Biol Chem 2004; 279(47): 49199-205. [257] Yang G, Wu L, Wang R. Pro-apoptotic effect of endogenous H2S on human aorta smooth muscle cells. FASEB J 2006; 20(3): 553-5. [258] Cario E, Rosenberg IM, Brandwein SL, Beck PL, Reinecker HC, Podolsky DK. Lipopolysaccharide activates distinct signaling pathways in intestinal epithelial cell lines expressing Toll-like receptors. J Immunol 2000; 164(2): 966-72. [259] Jeong SO, Pae HO, Oh GS, et al. Hydrogen sulfide potentiates interleukin1beta-induced nitric oxide production via enhancement of extracellular signalregulated kinase activation in rat vascular smooth muscle cells. Biochem Biophys Res Commun 2006; 345(3): 938-44. [260] Jarrar D, Kuebler JF, Rue LW, et al. Alveolar macrophage activation after trauma-hemorrhage and sepsis is dependent on NF-κB and MAPK/ERK mechanisms. Am J Physiol Lung Cell Mol Physiol 2002; 283(4): L799-805. [261] Yang G, Sun X, Wang R. Hydrogen sulfide-induced apoptosis of human aorta smooth muscle cells via the activation of mitogen-activated protein kinases and caspase-3. FASEB J 2004; 18(14): 1782-4. [262] Yong QC, Choo CH, Tan BH, Low CM, Bian JS. Effect of hydrogen sulfide on intracellular calcium homeostasis in neuronal cells. Neurochem Int 2010; 56(3): 50815. 216 [263] Lee SW, Hu YS, Hu LF, et al. Hydrogen sulphide regulates calcium homeostasis in microglial cells. Glia 2006; 54(2): 116-24. [264] Tamizhselvi R, Sun J, Koh YH, Bhatia M. Effect of hydrogen sulfide on the phosphatidylinositol 3-kinase-protein kinase B pathway and on caerulein-induced cytokine production in isolated mouse pancreatic acinar cells. J Pharmacol Exp Ther 2009; 329(3): 1166-77. [265] Russell JA, Boyd J, Nakada T, Thair S, Walley KR. Molecular mechanisms of sepsis. Contrib Microbiol 2011; 17: 48-85. [266] Varisco BM. The pharmacology of acute lung injury in sepsis. Adv Pharmacol Sci 2011; doi: 10.1155/2011/254619. [267] Wheeler AP, Bernard GR. Acute lung injury and the acute respiratory distress syndrome: a clinical review. Lancet 2007; 369(9572): 1553-64. [268] Ricciotti E, FitzGerald GA. Prostaglandins and inflammation. Arterioscler Thromb Vasc Biol 2011; 31(5): 986-1000. [269] Ohara M, Sawa T, Kurahashi K, et al. Induction of cyclooxygenase-2 in alveolar macrophages after acid aspiration: selective cyclooxygenase-2 blockade reduces interleukin-6 production. Anesthesiology 1998; 88(4): 1014-22. [270] Gust R, Kozlowski JK, Stephenson AH, Schuster DP. Role of cyclooxygenase2 in oleic acid-induced acute lung injury. Am J Respir Crit Care Med 1999; 160(4): 1165-70. [271] Ejima K, Layne MD, Carvajal IM, et al. Cyclooxygenase-2-deficient mice are resistant to endotoxin-induced inflammation and death. FASEB J 2003; 17(10): 13257. [272] Li B, Li YM, Li X, et al. COX-2 inhibition improves immune system homeostasis and decreases liver damage in septic rats. J Surg Res 2009; 157(1): 43-7. 217 [273] Shoup M, He LK, Liu H, Shankar R, Gamelli R. Cyclooxygenase-2 inhibitor NS-398 improves survival and restores leukocyte counts in burn infection. J Trauma 1998; 45(2): 215-20; discussion 20-1. [274] Sio SW, Ang SF, Lu J, Moochhala S, Bhatia M. Substance P Upregulates Cyclooxygenase-2 and Prostaglandin E Metabolite by Activating ERK1/2 and NF-κB in a Mouse Model of Burn-Induced Remote Acute Lung Injury. J Immunol 2010; 185(10): 6265-76. [275] Koon HW, Zhao D, Zhan Y, Rhee SH, Moyer MP, Pothoulakis C. Substance P stimulates cyclooxygenase-2 and prostaglandin E2 expression through JAK-STAT activation in human colonic epithelial cells. J Immunol 2006; 176(8): 5050-9. [276] Gallicchio M, Benetti E, Rosa AC, Fantozzi R. Tachykinin receptor modulation of cyclooxygenase-2 expression in human polymorphonuclear leucocytes. Br J Pharmacol 2009; 156(3): 486-96. [277] Gallicchio M, Rosa AC, Benetti E, Collino M, Dianzani C, Fantozzi R. Substance P-induced cyclooxygenase-2 expression in human umbilical vein endothelial cells. Br J Pharmacol 2006; 147(6): 681-9. [278] Chen Y, Chen P, Hanaoka M, Droma Y, Kubo K. Enhanced levels of prostaglandin E2 and matrix metalloproteinase-2 correlate with the severity of airflow limitation in stable COPD. Respirology 2008; 13(7): 1014-21. [279] Lappi-Blanco E, Kaarteenaho-Wiik R, Maasilta PK, Anttila S, Paakko P, Wolff HJ. COX-2 is widely expressed in metaplastic epithelium in pulmonary fibrous disorders. Am J Clin Pathol 2006; 126(5): 717-24. [280] Carpagnano GE, Spanevello A, Palladino GP, et al. Cigarette smoke and increased COX-2 and survivin levels in exhaled breath condensate of lung cancer patients: how hot is the link? Lung Cancer 2009; 67(1): 108-13. 218 [281] Hahn EL, Tai HH, He LK, Gamelli RL. Burn injury with infection alters prostaglandin E2 synthesis and metabolism. J Trauma 1999; 47(6): 1052-7. [282] Futaki N, Takahashi S, Kitagawa T, Yamakawa Y, Tanaka M, Higuchi S. Selective inhibition of cyclooxygenase-2 by NS-398 in endotoxin shock rats in vivo. Inflamm Res 1997; 46(12): 496-502. [283] Attene-Ramos MS, Nava GM, Muellner MG, Wagner ED, Plewa MJ, Gaskins HR. DNA damage and toxicogenomic analyses of hydrogen sulfide in human intestinal epithelial FHs 74 Int cells. Environ Mol Mutagen 2010; 51(4): 304-14. [284] Hu LF, Pan TT, Neo KL, Yong QC, Bian JS. Cyclooxygenase-2 mediates the delayed cardioprotection induced by hydrogen sulfide preconditioning in isolated rat cardiomyocytes. Pflugers Arch 2008; 455(6): 971-8. [285] Piper PJ. Release and metabolism of prostaglandins in lung tissue. Pol J Pharmacol Pharm 1974; 26(1): 61-72. [286] Piper PJ, Vane JR, Wyllie JH. Inactivation of prostaglandins by the lungs. Nature 1970; 225(5233): 600-4. [287] Wyllie JH, Piper PJ, Vane JR. Fate of prostaglandins in the lungs. Br J Surg 1969; 56(8): 623. [288] Granstrom E, Hamberg M, Hansson G, Kindahl H. Chemical instability of 15keto-13,14-dihydro-PGE2: the reason for low assay reliability. Prostaglandins 1980; 19(6): 933-57. [289] Bothwell W, Verburg M, Wynalda M, Daniels EG, Fitzpatrick FA. A radioimmunoassay for the unstable pulmonary metabolites of prostaglandin E1 and E2: an indirect index of their in vivo disposition and pharmacokinetics. J Pharmacol Exp Ther 1982; 220(2): 229-35. 219 [290] Maclouf J, Grassi J, Pradelles P. Development of enzyme-immunoassay techniques for the measurement of eicosanoids. In: Prostaglandin and Lipid Metabolism in Radiation Injury; Walden TLJ, Hughes HN, Ed. Rockville, MD: Plenum Press 1987; pp. 355-64. [291] Ivanov AI, Scheck AC, Romanovsky AA. Expression of genes controlling transport and catabolism of prostaglandin E2 in lipopolysaccharide fever. Am J Physiol Regul Integr Comp Physiol 2003; 284(3): R698-706. [292] Duffield-Lillico AJ, Boyle JO, Zhou XK, et al. Levels of prostaglandin E metabolite and leukotriene E(4) are increased in the urine of smokers: evidence that celecoxib shunts arachidonic acid into the 5-lipoxygenase pathway. Cancer Prev Res (Phila) 2009; 2(4): 322-9. [293] Gross ND, Boyle JO, Morrow JD, et al. Levels of prostaglandin E metabolite, the major urinary metabolite of prostaglandin E2, are increased in smokers. Clin Cancer Res 2005; 11(16): 6087-93. [294] Padi SS, Jain NK, Singh S, Kulkarni SK. Pharmacological profile of parecoxib: a novel, potent injectable selective cyclooxygenase-2 inhibitor. Eur J Pharmacol 2004; 491(1): 69-76. [295] Talley JJ, Bertenshaw SR, Brown DL, et al. N-[[(5-methyl-3-phenylisoxazol-4yl)-phenyl]sulfonyl]propanamide, sodium salt, parecoxib sodium: A potent and selective inhibitor of COX-2 for parenteral administration. J Med Chem 2000; 43(9): 1661-3. [296] Teagarden DL, Sandeep N. Case study: parecoxib: a prodrug of valdecoxib. In: Prodrugs: Challenges and Rewards (Part 1: Biotechnology: Pharmaceutical Aspects); Stella VJ, Borchardt RT, Hageman MJ, Oliyai R, Maag H, Tilley JW, Ed. St. Louis, MO: Springer New York 2007; pp. 1335-46. 220 [297] Cuzzocrea S, Mazzon E, Sautebin L, et al. Protective effects of Celecoxib on lung injury and red blood cells modification induced by carrageenan in the rat. Biochem Pharmacol 2002; 63(4): 785-95. [298] Fukushima R, Alexander JW, Wu JZ, et al. Time course of production of cytokines and prostaglandin E2 by macrophages isolated after thermal injury and bacterial translocation. Circ Shock 1994; 42(3): 154-62. [299] Portanova JP, Zhang Y, Anderson GD, et al. Selective neutralization of prostaglandin E2 blocks inflammation, hyperalgesia, and interleukin production in vivo. J Exp Med 1996; 184(3): 883-91. [300] Rasley A, Marriott I, Halberstadt CR, Bost KL, Anguita J. Substance P augments Borrelia burgdorferi-induced prostaglandin E2 production by murine microglia. J Immunol 2004; 172(9): 5707-13. [301] Ishii I, Akahoshi N, Yamada H, Nakano S, Izumi T, Suematsu M. Cystathionine gamma-Lyase-deficient mice require dietary cysteine to protect against acute lethal myopathy and oxidative injury. J Biol Chem 2010; 285(34): 26358-68. 221 [...]... increasingly being established as a novel mediator of inflammation It has been demonstrated to play a pro-inflammatory role in animal models of local and systemic inflammation, including carrageenan-induced hindpaw edema [7], burn-induced acute lung injury (ALI) [8], caerulein-induced acute pancreatitis [9], lipopolysaccharide (LPS)-evoked endotoxemia [10], and cecal ligation and puncture (CLP)-induced... sequelae remain an important global healthcare problem for both scientists and clinicians and are a leading cause of morbidity and mortality in medical and surgical intensive care units (ICUs) Therefore, intense research on elucidating the cascades of mechanisms associated with pathogenesis of sepsis and potential preventive and therapeutic strategies are of great interest Nitric oxide (NO), carbon... Effect of PAG and capsazepine on protein expression and activity of COX-2 in septic lungs 168 Figure 6.3 Effect of NaHS or PAG and capsazepine on PGE2 metabolite production in septic lungs 170 Figure 6.4 Effect of NaHS and parecoxib on lung MPO activity, histopathological evaluation (hematoxylin and eosin staining) of lung polymorphonuclear leukocyte infiltration and injury, and pulmonary edema in sepsis. .. prophylactic PAG, and septic mice received therapeutic PAG 178 Figure 7.1 Flowchart summarizing the pro-neuroinflammatory role of H2S in polymicrobial sepsis 188 xv LIST OF ABBREVIATIONS 3-MST 3-mercaptopyruvate sulfurtransferase AC adenylyl cyclase AECOPD Acute exarcebation of chronic obsructive pulmonary disease ALI Acute lung injury ALT Alanine aminotransferase ANOVA Analysis of variance ARDS Acute... of PAG and capsazepine on lung and liver MPO activity in septic mice 95 Figure 3.10 Effect of PAG and capsazepine on lung and liver injury in septic mice 96 Figure 3.11 Effect of PAG and capsazepine on CLP-induced mortality in septic mice 97 xiii Figure 4.1 Effect of NaHS and capsazepine on lung and plasma SP levels, and lung PPT -A mRNA expression in septic mice 112 Figure 4.2 Effect of NaHS and capsazepine... SWS, Ang SF, Lu J, Moochhala SM, Bhatia M Substance P Upregulates Cyclooxygenase-2 and Prostaglandin E Metabolite by Activating ERK1/2 and NF-κB in a Mouse Model of Burn-Induced Remote Acute Lung Injury Journal of Immunology 2010; 185(10):6265-6276 ABSTRACT Ang SF, Moochhala SM, MacAry PA, Bhatia M Hydrogen Sulfide Regulates Transient Receptor Potential Vanilloid 1-Mediated Neurogenic Inflammation in Sepsis- Associated... hypothesized that H2S may modulate neuroinflammation in sepsis However, there has been little progress in understanding the potential interaction and involvement of both H2S and TRPV1 in the setting of sepsis Therefore, in the present study, we have investigated the potential role of H2S in instigating TRPV1-mediated neurogenic inflammation in a mouse model of polymicrobial sepsis Additionally, we have identified... MacAry PA, Bhatia M 2010 A Key Role of Substance P in Hydrogen Sulfide- Induced Neurogenic Inflammation in Sepsis- Associated Lung Injury 10th Annual Meeting of the Federation of Clinical Immunology Societies (FOCIS 2010), June 24-27, Boston, Massachusetts, USA Ang SF, Moochhala SM, MacAry PA, Bhatia M 2011 Hydrogen Sulfide Regulates Transient Receptor Potential Vanilloid 1-Mediated Neurogenic Inflammation. .. expression of adhesion molecules in the lung and liver of septic mice 120 Figure 4.6 Effect of NaHS and capsazepine on plasma ALT and AST activities, and pulmonary edema (measured as lung wet-to-dry weight ratio) in septic mice 122 Figure 4.7 Effect of PAG and capsazepine on lung and plasma SP levels, and lung PPT -A mRNA expression in septic mice 124 Figure 4.8 Effect of PAG and capsazepine on protein levels... distributions are not homogenous CBS appears to be the main H2S-synthesizing enzyme in the CNS and is highly expressed in liver, kidney, and the hippocampus and cerebellum in mammalian brain CSE is primarily responsible for H2S formation in the cardiovascular system and is predominantly found in the liver and in vascular and non-vascular smooth muscle, and at much lower levels, in small intestine and stomach of . gastrointestinal, genitourinary, and nervous systems. Besides acting as a potent vasodilator and an atypical neuromodulator, H 2 S is increasingly being established as a novel mediator of inflammation. . Effect of NaHS and capsazepine on mRNA expression of adhesion molecules in the lung and liver of septic mice 120 Figure 4.6 Effect of NaHS and capsazepine on plasma ALT and AST activities, and. Effect of PAG and capsazepine on mRNA expression of adhesion molecules in the lung and liver of septic mice 132 Figure 4.12 Effect of PAG and capsazepine on plasma ALT and AST activities, and

Ngày đăng: 10/09/2015, 08:32

Từ khóa liên quan

Mục lục

  • Acknowledgements

  • Summary

  • List of Tables

  • List of Figures

  • List of Abbreviations

  • Publications

  • Chapter I Introduction

    • 1.1 General overview

    • 1.2 H2S

    • 1.2.1 Chemical properties of H2S

    • 1.2.2 H2S toxicity

    • 1.2.3 Biosynthesis of H2S

    • 1.2.4 Metabolism of H2S

    • 1.2.5 Biological roles of H2S

    • 1.2.5.1 Roles of H2S in CNS

    • 1.2.5.1.1 Physiological roles of H2S in CNS

    • 1.2.5.1.2 H2S in CNS diseases

    • 1.2.5.2 Roles of H2S in cardiovascular system

    • 1.2.5.2.1 Physiological roles of H2S in cardiovascular system

    • 1.2.5.2.2 H2S in cardiovascular diseases

    • 1.2.5.3.2 H2S in gastrointestinal diseases

Tài liệu cùng người dùng

Tài liệu liên quan