Cytoskeleton regulators in the control of corneal fibrosis

147 224 0
Cytoskeleton regulators in the control of corneal fibrosis

Đang tải... (xem toàn văn)

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

Thông tin tài liệu

i Acknowledgements I would like to express my sincere gratitude to Professor Roger W. Beuerman for his supervision and guidance throughout this project. I am grateful for his comments and advice. I would like to thank all the staff of SERI especially Sia-Wey Yeo, Wing-Sam Lee, Jennifer Ng, Queenie Tan, Candice Ho, Jia Lin, Wan-e Lim and Dr. Amutha Veluchamy Barathi, who assisted me in many ways. My work was supported by the grant: National Research Council of Singapore grant (R739/23/2010) and Singhealth Foundation grant (R835/30/2011). I would like to thank my family for their love and support throughout my endeavours. ii TABLE OF CONTENTS CHAPTER I. INTRODUCTION…….………………………………………………. 1 Cornea structure…………………………………………………… ……… 1.1.1 Corneal stroma structureh …………………………………………… .……. 1.1.2 Corneal stroma keratocytes…………………………………………………. 1.1.3 Cytoskeleton in biology………………………………………………… …. 1.1.4 Cytoskeleton regulators in biology…………………………….……………. 1.2 Corneal fibrosis as an adverse outcome…………………… ………………. 1.3 Corneal stroma keratocytes in corneal fibrosis……………………………… 1.4 Mediators trigger corneal fibrosis……………………… …………………. 1.4.1 TGF-β1 induced fibrosis……………………………………………………. 1.4.2 TGF-β1 induced corneal fibrosis………………………… .………………. 1.5 Cytoskeleton in fibrosis……………………………………………….……. 1.5.1 Cytoskeleton regulators in fibrosis…………………………………………. 1.6 Current treatment of corneal fibrosis………………………….……………. 10 1.7 Answering the Experimental Questions……………………………………. 12 1.8 Hypothesis…………………………………………………………….……. 12 1.8.1 Specific aim…………………………………………………………………. 13 CHAPTER II. MATERIAL AND METHODS…………………………………… 14 2.1 Ethics……………………………………………………………………… . 14 2.2 Anterior Keratectomy Procedure……………………………………………. 14 2.3 Animal Experimental Groups………………………………………… …… 15 2.4 Gold-Chloride Procedure………………………………………………….…. 16 iii 2.5 Cytoskeleton Regulators RT² Profiler™ PCR Array………………………… 16 2.6 Moesin siRNA in vivo delivery…………………………………… ………. 17 2.7 List of laboratory techniques………………………………………… ……. 18 2.7.1 Preparation of tissue…………………………………………………………. 18 2.7.2 Use of exogenous protein……………………………………………………. 18 2.7.3 Histological Evaluation …………………………….………………………. 19 2.7.4 Immunochemistry……………………………………………………… …. 21 2.7.5 Imaging…………………………………………………………………… 22 2.7.6 RNA Extraction and quantification……………………………… ………. 22 2.7.7 Reverse transcription and Polymerase chain reaction…………………… 24 2.7.8 Real time RT-PCR………………………………………………………… 25 2.7.9 Total protein extraction……………………………………………………. 27 2.7.10 BCA protein assay………………………………………………………… 28 2.7. 11 Western blot analysis……………………………………………………… 28 2.8 30 Statistical analysis…………………………………………………………. Chapter III. TGF-β1-induced corneal fibrosis…………………………………… 31 3.1 Phenotypic changes of corneal stroma keratocytes in response to the anterior keratectomy…………………………………………………………………. 31 3.2 Effect of various dosing regimes of topical application of TGF-β1 on the transformation of corneal keratocytes-to-myofibroblasts in corneal fibrosis in vivo……………………………………. ……………………………………. 33 3.3 TGF-β1 induced transformation of corneal stroma keratocytes-tomyofibroblasts in the corneal fibrosis……………………………….………. 39 Up-regulation of cytoskeleton regulators in corneal fibrosis in vivo……… 41 3.4 iv Chapter IV. Moesin and fibrosis………………………………………………………. 46 4.1 Moesin functions in cytoskeleton……………… .……………………………. 46 4.2 Moesin in fibrosis…………………………………………………… .………. 46 4.3 Co-localization of moesin and α-SMA within corneal stroma keratocytes in corneal fibrosis………………………………………………………………… 47 4.4 Characterization of moesin expression in the corneal fibrosis………………… 49 4.5 Role of moesin in the corneal fibrosis in vivo…………………………………. 51 4.5.1 In vivo delivery of moesin siRNA into the cornea after an anterior keratectomy…………………………………………………………………… 51 4.5.2 The transformation of corneal stroma keratocytes-to-myofibroblasts in the corneal fibrosis is moesin-dependent…………………………………………. 56 4.5.3 Corneal haze development in the injured cornea……………………………… 60 Chapter V. Moesin and signaling in corneal fibrosis 63 5.1 Introduction to Smad signaling in fibrosis……………………………………. 63 5.2 Effect of TGF-β1 on the activation of Smad and Smad in the corneal fibrosis………………………………………………………………………… 64 5.3 Effect of moesin on TGF-β1-stimulated activation of Smad2 and Smad3 in corneal fibrosis…………………………………………………………………. 68 Chapter VI. Discussion………………………. 72 6.1 Summary of findings………………………………………………………… 72 6.1.1 Transformation of corneal stroma keratocytes to myofibroblasts in corneal fibrosis…………………………………………………………………………. 72 6.1.2 Effect of TGF-β1 on corneal fibrosis……………………….…………………. 73 6.1.3 Role of moesin in the corneal fibrosis in vivo…………………………………. 73 6.1.4 Signaling pathway relevant to the corneal fibrosis regulated by moesin in Vivo…………………………………………………………………………… 75 6.2 Limitations…………………………………………………………………… 77 v 6.3 Clinical application……………………………………………………………. 79 6.4 Future studies………………………………………………………………… 81 Chapter VII. References……………………………………………………………. 85 Chapter VIII. Appendices………………………………………………………… 131 Appendix A: Experimental animal groups……………………………………………. 132 Appendix B: Moesin siRNA sequences………………………………………………. 132 Appendix C: Antibodies………………………………………………………………. 132 Appendix D: PCR primers……………………………………………………………. 133 Chapter X. Supplementary…………………………………………………………… 134 A. Grants: ……………………………………………………………………………… 134 B. Patents………………………………………………………………………………. 134 C. Presentations at Conferences……………………………………………………… 134 D. Publications…………………………………………………………………………. 136 vi SUMMARY The avascular cornea accounts for about two-thirds of the total refractive power of the eye, therefore maintenance of corneal transparency is critical for focusing light onto the retina. The impact on vision is highlighted by the finding that corneal scar as a contributor to “corneal blindness” is the third leading cause of blindness world-wide. Corneal scar may develop as sequel to infections and a wide spectrum of corneal stromal injuries such as refractive surgery (LASIK, etc.). Currently, corneal transplantation is the major treatment regime for corneal scar. To develop more specific medical therapies or as an adjunct to surgery to prevent scar formation, the regulation of corneal scar must be understood. Fibrosis is the cellular process that leads to the formation of scar. A strong association of fibrosis with “cytoskeleton regulators” suggests this class of proteins that links the intracellular cytoskeleton with the extracellular environment may be a target for therapeutic development; however, as this is a large group of proteins, evidence for involvement of specific members of this class of proteins in the corneal fibrosis has not been developed. Recently, interest in the ERM (ezrin/radixin/moesin) family members has shown that these proteins are the organizer of membrane domains and act as links to the cytoskeleton as well as signalling pathways involved in many cellular processes. Cytoskeleton disruption agents have been shown to prevent fibrosis may involve this family of proteins. Our clinical target is the early phase of corneal wounding, before an opaque corneal scar forms. In response to corneal injury, the transformation of corneal stroma keratocytes-to-myofibroblasts is predominently responsible for the corneal fibrosis. vii To define the in vivo role of specific cytoskeleton regulators in mediating the corneal fibrosis, a mouse model with corneal fibrosis stimulated by the anterior keratectomy was established. TGF-β1 was topically applied to accelerate the corneal fibrosis process since it activates corneal stroma keratocytes to a myofibroblast phenotype expressing α-SMA accompanied by altered expression patterns of ECM components. The main results of the present study are that (1) in the corneal fibrosis, moesin was identified as the most highly induced gene among the 84 cytoskeleton regulator genes using cytoskeleton regulators RT² Profiler™ PCR array (Chapter III 3.4); (2) The upregulation of moesin in the corneal fibrosis was confirmed by RT-PCR (Chapter III 3.4) and western blot (Chapter IV 4.4) in a time-dependent manner; (3) the appearance of myofibroblasts was analyzed by examining α-SMA expression.Dual immunofluorescent staining showed that moesin colocalized with α-SMA within corneal stroma keratocytes in the corneal fibrosis (Chapter IV 4.3); (4) Up-regulation of α-SMA and moesin in the corneal fibrosis was reduced by moesin siRNA (Chapter IV 4.5.1 and 4.5.2); (5) Moesin siRNA reduced corneal opacification in corneal fibrosis (Chapter IV 4.5.3); (6) Activation of Smad and Smad in corneal fibrosis was reduced by moesin siRNA (Chapter V 5.3). In conclusion, moesin siRNA decreased the transformation of corneal stroma keratocytes-to-myofibroblasts in the corneal fibrosis, as defined by the expression of α-SMA, through the reduction of activated forms of Smad and Smad 3. Moesin may be a potential drug target for inhibiting corneal fibrosis and details of moesin-related signaling pathways would be critical for understanding corneal fibrosis. viii LIST OF TABLES 1. Experimental animal groups…………………………………………… 133 2. List of moesin siRNA sequence……………………………………… .133 3. List of antibodies……………………………………………………… 132 4. List of PCR primers…………………………………………………… .132 5. Various dosing regimes for topical application of TGF-β1 on the corneal stroma after the anterior keratectomy…………………………… 35 ix LIST OF FIGURES Histological analysis of corneal wound healing after an anterior keratectomy……………………………………………………………………20 Phenotypic changes of corneal stromal keratocytes in response to the anterior keratectomy .………………………………………… 32 Effect of various dosing regimes for TGF-β1 on the transformation of corneal keratocytes-to-myofibroblasts, as defined by the expression of α-SMA in the corneal stroma after the anterior keratectomy 35 Keratocyte-to-myofibroblast transformation induced by topical application of TGF-β1, as defined by α-SMA expression .40 Results for the response of cytoskeleton regulators to topical application of TGF-ß1 after an anterior keratectomy at PO day analyzed by RT² Profiler PCR-array ……….………………………………………………42 RT-PCR quantification of mRNA levels for cytoskeleton regulators induced by topical application of TGF-ß1 following an anterior keratectomy .44 Dual immunofluorescent staining demonstrated that moesin co-localized with α-SMA within corneal stroma keratocytes in the corneal fibrosis…………… 48 x Up-regulation of moesin in the corneal fibrosis……… ……………………50 Distribution of fluorescein–labeled moesin siRNA after in vivo delivery of moesin siRNA by iontophoresis into the cornea…………….52 10 Effect of moesin siRNA on the expression of moesin in the corneal fibrosis ……………………………………………………………….54 11 Effect of moesin siRNA on the transformation of corneal keratocytes-to-myofibroblasts in the corneal fibrosis ……………….……… 58 12 In vivo slit lamp macroscopic observation of the cornea in group 2-AK and group 5-AKTSi……………………………………………………………61 13 Effect of TGF-β1 on Smad activation …………………………………… 65 14 Effect of TGF-β1 on Smad activation ………………………………… …67 15 Effect of moesin on Smad activation …………………… ……………….69 16 Effect of moesin on Smad activation ………………….………………… 70 121 Sharma A, Mehan MM, Sinha S, Cowden JW, Mohan RR. Trichostatin a inhibits corneal haze in vitro and in vivo. Invest Ophthalmol Vis Sci. 2009;50:2695-701. Shah M, Foreman dM, Ferguson MWJ. Neutralising antibody to TGF-â1,2 reduces cutaneous scarring in adult rodents. J Cell Sci. 1994;106:1137–1157. Shah M, Foreman DM, Furgeson MWJ. Neutralisation of TGF-â1 and TGF-â2 or exogenous addition of TGF-â3 to cutaneous rat wounds reduces scarring. J Cell Sci. 1995;108:985–1002. Shi-Wen X, Chen Y, Denton CP, Eastwood M, Renzoni EA, Bou-Gharios G, Pearson JD, Dashwood M, du Bois RM, Black CM, Leask A, Abraham DJ. Endothelin-1 promotes myofibroblast induction through the ETA receptor via a rac/phosphoinositide 3-kinase/Akt-dependent pathway and is essential for the enhanced contractile phenotype of fibrotic fibroblasts. Mol Biol Cell. 2004;15: 2707–2719. Silacci P, Mazzolai L, Gauci C, Stergiopulos N, Yin HL, Hayoz D. Gelsolin superfamily proteins: key regulators of cellular functions. Cell Mol Life Sci. 2004;61(19-20):2614-23. Sime PJ, Xing Z, Foley R, Graham FL, Gauldie J. Transient gene transfer and expression in the lung. Chest. 1997a;111(6 Suppl):89S-94S. Sime PJ, Xing Z, Graham FL, Csaky KG, Gauldie J. Adenovectormediated gene transfer of active transforming growth factor beta1 induceds prolonged severe fibrosis in rat lung. J Clin Invest 1997b; 100: 768–76. Singer AJ, Clark RA. 1999;341(10):738-46. Cutaneous wound healing. N Engl J Med. 122 Singer II, Kawka DW, Kazazis DM, Clark RA. In vivo co-distribution of fibronectin and actin fibers in granulation tissue: immunofluorescence and electron microscope studies of the fibronexus at the myofibroblast surface. J. Cell Biol. 1984;98:2091–2106. Skalli O, Gabbiani G. The Biology of the Myofibroblast. Relationship to Wound Contraction and Fibrocontractive Diseases. In: The molecular and cellular biology of wound repair, CLARK R.A.F. and HENSON P.M., eds, Plenum Press, New York, 1988; 373-402. Smelser GK, Ozanic V. New concepts in anatomy and histology of the cornea. In: King JH, McTigue JW, eds. The Cornea. Washington, DC: Butterworths. 1965:120. Smith JW, Christie KN, Frame J. Desmosomes, cilia and acanthosomes associated with keratocytes. J Anat. 1969; 105:383-392. Soini Y, Satta J, Maatta M, Autio-Harmainen H. Expression of MMP2, MMP9, MT1-MMP, TIMP1, and TIMP2 mRNA in valvular lesions of the heart. J Pathol. 2001; 194: 225–231. Soutschek J, Akinc A, Bramlage B, Charisse K, Constien R, Donoghue M, Elbashir S, Geick A, Hadwiger P, Harborth J, John M, Kesavan V, Lavine G, Pandey RK, Racie T, Rajeev KG, Röhl I, Toudjarska I, Wang G, Wuschko S, Bumcrot D, Koteliansky V, Limmer S, Manoharan M, Vornlocher HP. Therapeutic silencing of an endogenous gene by systemic administration of modified siRNAs. Nature. 2004;432:173-8. 123 Sporn MB, Roberts AB. Transforming growth factor-β: Recent progress and new challenges. J. Cell Biol. 1992;119:1017– 1021. Stabile H, Carlino C, Mazza C, Giliani S, Morrone S, Notarangelo LD, Notarangelo LD, Santoni A, Gismondi A. Impaired NK-cell migration in WAS/XLT patients: role of Cdc42/WASp pathway in the control of chemokineinduced beta2 integrin high-affinity state. Blood. 2010 Apr 8;115(14):2818-26. Stramer BM, Austin JS, Roberts AB, Fini ME. Selective reduction of fibrotic markers in repairing corneas of mice deficient in Smad3. J Cell Physiol. 2005;203:226-32. Sun H, Kwiatkowska K, Wooten DC, Yin HL. Effects of CapG overexpression on agonistinduced motility and second messenger generation. J. Cell Biol. 1995;129:147-56. Sun HQ, Yamamoto M, Mejillano M, Yin HL: Gelsolin, a multifunctional actin regulatory protein. J Biol Chem. 1999; 274:33179–33182, Takahashi E, Nagano O, Ishimoto T, Yae T, Suzuki Y, Shinoda T, Nakamura S, Niwa S, Ikeda S, Koga H, Tanihara H, Saya H. Tumor necrosis factor-alpha regulates transforming growth factor-beta-dependent epithelial-mesenchymal transition by promoting hyaluronan-CD44-moesin interaction. J Biol Chem. 2010;285:4060-73. Takeuchi K, Sato N, Kasahara H, Funayama N, Nagafuchi A, Yonemura S, Tsukita S, Tsukita S. Perturbation of cell adhesion and microvilli formation by antisense oligonucleotides to ERM family members. J Cell Biol. 1994a;125: 1371–1384. 124 Takeuchi K, Kawashima A, Nagafuchi A, Tsukita S. Structural diversity of band 4.1 superfamily members. J Cell Sci. 1994b;107:1921-8. Tamm ER, Siegner A, Baur A, Lutjen-Drecoll E. Transforming growth factor-β1 induces α-smooth muscle-actin expression in cultured human and monkey trabecular meshwork. Exp. Eye Res. 1996;62:389– 397. Tandon A, Tovey JC, Sharma A, Gupta R, Mohan RR. Role of transforming growth factor Beta in corneal function, biology and pathology. Curr Mol Med. 2010;10:565-78. Tegtmeyer S, Reichl S, Muller-Goymann CC. Cultivation and characterization of a bovine in vitro model of the cornea. Pharmazie. 2004;59:464–471. Thannickal VJ, Lee DY, White ES, Cui Z, Larios JM, Chacon R, Horowitz JC, Day RM, Thomas PE. Myofibroblast differentiation by transforming growth factor-beta1 is dependent on cell adhesion and integrin signaling via focal adhesion kinase. J Biol Chem. 2003;278:12384–12389. Thannickal VJ, Toews GB, White ES, Lynch JP, III, Martinez FJ. Mechanisms of pulmonary fibrosis. Annu Rev Med. 2004;55:395–417. Thornton I, Xu M, Krueger RR. Comparison of standard (0.02%) and low dose (0.002%) mitomycin C in the prevention of corneal haze following surface ablation for myopia. J Refract Surg. 2008;24:S68–S76. Tomasek JJ, Haaksma CJ, Eddy RJ, Vaughan MB. Fibroblast contraction occurs on release of tension in attached collagen lattices: dependency on an organized actin cytoskeleton and serum. Anat. Rec. 1992;232:359–368. 125 Tomasek JJ, Gabbiani G, Hinz B, Chaponnier C, Brown RA. Myofibroblasts and mechano-regulation of connective tissue remodelling. Nat Rev Mol Cell Biol. 2002;3:349–363. Treanor B, Depoil D, Bruckbauer A, Batista FD. Dynamic cortical actin remodeling by ERM proteins controls BCR microcluster organization and integrity. J Exp Med. 2011;208:1055-68. Tsukita S, Yonemura S. Cortical actin organization: lessons from ERM (ezrin/radixin/moesin) proteins. J Biol Chem. 1999;274: 34507–34510. Tuori A, Virtanen I, Aine E, Uusitalo H. The expression of tenascin and fibronectin in keratoconus, scarred and normal human cornea. Graefes Arch Clin Exp Ophthalmol 1997;235:222–229. Turner CE: Molecules in focus Paxillin. Int J Biochem Cell Biol. 1998; 30: 955– 959, Turner CE: Paxillin interactions. J Cell Sci. 2000; 113: 4139–4140. Turunen O, Wahlström T, Vaheri A. Ezrin has a COOH-terminal actin-binding site that is conserved in the ezrin protein family. J Cell Biol. 1994;126:1445-53. Ueno H, Sakamoto T, Nakamura T, Qi Z, Astuchi N, Takeshita A. A soluble transforming growth factor beta receptor expressed in muscle prevents liver fibrogenesis in rats. Hum Gene Ther 2000; 11: 33–42. Vaheri A, Carpén O, Heiska L, Helander TS, Jääskeläinen J, MajanderNordenswan P, Sainio M, Timonen T, Turunen O. The ezrin protein family: membrane-cytoskeleton interactions and disease associations. Curr Opin Cell Biol. 1997;9:659-66. 126 van Beurden HE, Snoek PA, Von den Hoff JW, Torensma R, Maltha JC, Kuijpers-Jagtman AM. Dynamic protein expression patterns during intraoral wound healing in the rat. Eur J Oral Sci. 2005;113:153-8. Van den Berg TJ. Importance of pathological intraocular light scatter for visual disability. Doc Ophthalmol. 1986;61:327–333. Vaughan MB, Howard EW, Tomasek JJ. Transforming growth factor-beta1 promotes the morphological and functional differentiation of the myofibroblast. Exp Cell Res. 2000;257: 180–189. Verma A, Guha S, Wang H, Fok JY, Koul D, Abbruzzese J, Mehta K. Tissue transglutaminase regulates focal adhesion kinase/AKT activation by modulating PTEN expression in pancreatic cancer cells. Clin Cancer Res. 2008 ;14:19972005. Vi L, de Lasa C, DiGuglielmo GM, Dagnino L. Integrin-linked kinase is required for TGF-β1 induction of dermal myofibroblast differentiation. J Invest Dermatol. 2011;131:586-93. Vittal R, Horowitz JC, Moore BB, Zhang H, Martinez FJ, Toews GB, Standiford TJ, Thannickal VJ. Modulation of prosurvival signaling in fibroblasts by a protein kinase inhibitor protects against fibrotic tissue injury. Am J Pathol. 2005;166:36775. Wade, R, Bohl, J, Vande Pol, S: Paxillin null embryonic stem cells are impaired in cell spreading and tyrosine phosphorylation of focal adhesion kinase. Oncogene 2002;21: 96–107. 127 Walker GA, Masters KS, Shah DN, Anseth KS, Leinwand LA. Valvular myofibroblast activation by transforming growth factor-beta: implications for pathological extracellular matrix remodeling in heart valve disease. Circ Res. 2004; 95: 253–260. Wamhoff BR, Bowles DK, Owens GK. Excitation-transcription coupling in arterial smooth muscle. Circ. Res. 2006;98:868–878. Wang J, Chen H, Seth A, McCulloch CA. Mechanical force regulation of myofibroblast differentiation in cardiac fibroblasts. Am. J. Physiol. Heart Circ. Physiol. 2003; 285:H1871–H1881. Wang Q, Wang Y, Hyde DM, Gotwals PJ, Koteliansky VE, Ryan ST. Reduction of bleomycin induced lung fibrosis by transforming growth factor soluble receptor in hamsters. Thorax. 1999; 54: 805–12. Weeds AG, Gooch J, Hawkins M, Pope B, Way M. Role of actin-binding proteins in cytoskeletal dynamics. Biochem Soc Trans. 1991;19:1016-20. Weimar, V. Healing processes in the cornea. In The Transparency of the Cornea (ed. S. Duke-Elder and E.S.Perkins), Blackwell Scientific Publications: Oxford, England. 1960. 111-124. Westermann S, Weber K. Post-translational modifications regulate microtubule function. Nat. Rev. Mol. Cell Biol. 2003; 4:938-47. Whyte M. Gelsolin in idiopathic pulmonary fibrosis: a new target supports a central role for epithelial injury in disease pathogenesis. Thorax. 2009;64:461-2. 128 Wilson SE, Mohan RR, Mohan RR, Ambrosio R Jr, Hong J, Lee J. The corneal wound healing response: cytokine-mediated interaction of the epithelium, stroma, and inflammatory cells. Progress in Retinal and Eye Research. 2001; 20: 625–637. Wilson SE. Analysis of the keratocyte apoptosis, keratocyte proliferation, and myofibroblast transformation responses after photorefractive keratectomy and laser in situ keratomileusis. Transactions of the American Ophthalmological Society 2002; 100: 411–433. Williams K, Ghosh R, Giridhar PV, Gu G, Case T, Belcher SM, Kasper S. Inhibition of stathmin1 accelerates the metastatic process. Cancer Res. 2012 Oct 15;72(20):5407-17. Witke, W, Sharpe, AH, Hartwig, JH, Azuma, T, Stossel, TP, Kwiatkowski, DJ: Hemostatic, inflammatory, and fibroblast responses are blunted in mice lacking gelsolin. Cell. 1995; 81: 41–51, Wrighton KH, Lin X, Yu PB, Feng XH. Transforming Growth Factor {beta} Can Stimulate Smad1 Phosphorylation Independently of Bone Morphogenic Protein Receptors. J Biol Chem. 2009;284:9755-63 Wu J, Ma B, Yi S, Wang Z, He W, Luo G, Chen X, Wang X, Chen A, Barisoni D. Gene expression of early hypertrophic scar tissue screened by means of cDNA microarrays. J Trauma. 2004;57:1276-86. Wu KY, Hong SJ, Huang HT, Lin CP, Chen CW. Toxic effects of mitomycin-C on cultured corneal keratocytes and endothelial cells. J Ocul Pharmacol Ther. 1999;15:401–411. 129 Wu M, McClellan SA, Barrett RP, Zhang Y, Hazlett LD. Beta-defensins and together promote resistance to Pseudomonas aeruginosa keratitis. J Immunol. 2009a;183:8054-60. Wu M, McClellan SA, Barrett RP, Hazlett LD. β-Defensin-2 promotes resistance against infection with P. aeruginosa. J Immunol. 2009b;182: 1609-1616. Wynn TA.,Common and unique mechanisms regulate fibrosis in various fibroproliferative diseases. J. Clin. Invest. 2007;117:524-9. Xia H, Nho RS, Kahm J, Kleidon J, Henke CA. Focal adhesion kinase is upstream of phosphatidylinositol 3-kinase/Akt in regulating fibroblast survival in response to contraction of type I collagen matrices via a beta integrin viability signaling pathway. J Biol Chem. 2004;279:33024-34. Yin HL. Gelsolin: calcium- and polyphosphoinositide-regulated actin-modulating protein. BioEssays. 1987;7:176-179. Yokozeki M, Moriyama K, Shimokawa H, Kuroda T. Transforming growth factor-beta modulates myofibroblastic phenotype of rat palatal fibroblasts in vitro. Exp Cell Res. 1997;231:328-36. Yonemura S, Nagafuchi A, Sato N, Tsukita S. Concentration of an integral membrane protein, CD43 (leukosialin, sialophorin), in the cleavage furrow through the interaction of its cytoplasmic domain with actin-based cytoskeletons. J Cell Biol. 1993;120:437-49. Yonemura S, Hirao M, Doi Y, Takahashi N, Kondo T, Tsukita S, Tsukita S. Ezrin/radixin/moesin (ERM) proteins bind to a positively charged amino acid 130 cluster in the juxta-membrane cytoplasmic domain of CD44, CD43, and ICAM-2, J. Cell. Biol.1998;140:885–895. Zhang HY, Gjaraee-Kermani M, Kaemiol S, Phan S.H. Lung fibroblast alpha smooth muscle actin expression and contractile phenotype in bleomycin-induced pulmonary fibrosis, Am. J. Pathol. 1996;148;527–537. Zhang HY, Phan SH. Inhibition of myofibroblast apoptosis by transforming growth factor _1. Am. J. Respir. Cell Mol. Biol. 1999;21:658–665. Zhang K, Rekhter MD, Gordon D, Phan SH. Myofibroblasts and their role in lung collagen gene expression during pulmonary fibrosis. A combined immunohistochemical and in situ hybridization study. Am J Pathol. 1994;145:114–125. Zhao J, Shi W, Wang YL, Chen H, Bringas P, Jr., Datto MB, Frederick JP, Wang XF, Warburton D. Smad3 deficiency attenuates bleomycin-induced pulmonary fibrosis in mice. Am J Physiol Lung Cell Mol Physiol. 2002;282: L585–L593. Zhu HY, Riau AK, Beuerman RW. Epithelial microfilament regulators show regional distribution in mouse conjunctiva. Mol Vis. 2010;16:2215-24. Zieske JD, Guimaraes SR, Hutcheon AE. Kinetics of keratocyte proliferation in response to epithelial debridement. Experimental Eye Research 2001; 72: 33–39. 131 Chapter VIII. Appendices Appendix A: Experimental animal groups Appendix B: Moesin siRNA sequence Appendix C: Antibodies Appendix D: PCR primers 132 Appendix A: Experimental animal groups Appendix B: Moesin siRNA sequences Appendix C: Antibodies 133 Appendix D: PCR primers 134 Chapter X. Supplementary A. Grants: 1. 2010 Singhealth Foundation Grant. Recipient of the grant: Zhu Hong-Yuan. 2. 2010 Association for Research in Vision and Ophthalmology (ARVO) International Annual Meeting-International Travel Grant. Recipient of the grant: Zhu Hong-Yuan. 3. 2011 National Medical Research Council Core Grant (Singapore Eye Research Institute Pilot Grant). Recipient of the grant: Zhu Hong-Yuan. 4. 2012 DUKE-NUS Khoo Scholar Grant. Recipient of the grant: Zhu HongYuan. B. Patents: 1. Moesin a new antifibrosis target in the cornea US provisional patent US Application N061/613, 633, March 21,2012 Zhu Hong-Yuan, RW Beuerman 2. High mobility group box protein and anti-inflammaotry target for postsurgical inflammation. 2012 Zhu Hong-Yuan, RW Beuerman C. Presentations at Conferences 1. Zhu Hong-Yuan, RW Beuerman. Molecular Characteristics and Regulation of Conjunctival Epithelial Cell Replacement. 17th Singapore General Hospital Scientific Congress, 2008. (Young Investigator Award, Merit). 135 2. Zhu Hong-Yuan, RW Beuerman. Integrin signaling pathways involved in the conjunctiva stem cell. 2009, 18th Singapore General Hospital scientific congress. (Young Investigator Award, Merit). 3. Zhu Hong-Yuan, Riau AK, RW Beuerman. Epithelial microfilament regulators show regional distribution in mouse conjunctiva. 2009, USA ARVO. Florida. 4. Zhu Hong-Yuan, RW Beuerman. Betamethasone and Ketorolac used to control postsurgical intraocular inflammation in the eye have different molecular target in the inflammatory pathway. 2010, International Singapore Symposium of Immunology. 5. Zhu Hong-Yuan, RW Beuerman. Mouse Model of Postsurgical Intraocular Inflammation. 2010, USA ARVO. Florida. (Travel grant) 6. Zhu Hong-Yuan, RW Beuerman. Betamethasone and Ketorolac used to control postsurgical intraocular inflammation in the eye have different molecular target in the inflammatory pathway. 2010, Singhealth Duke-NUS scientific congress, Singapore. (Allied Health Best Paper Award, Merit) 7. Zhu Hong-Yuan, SW Yeo, RW beuerman. Therapeutic Targets for PostOperative Intra-Ocular Inflammation. 2011, Asia Arvo, Singapore 8. Zhu Hong-Yuan, SW Yeo, RW beuerman. Cytoskeleton Regulators in Corneal Fibrosis. 19th Singapore General Hospital Scientific Congress, 2011 (Young Investigator Award, Merit). 136 D. Publications 1. Moesin as a key regulator in corneal fibrosis. Zhu Hong Yuan, SW Yeo, Beuerman RW. The Ocular Surface. 2012. In press. 2. Epithelial microfilament regulators show regional distribution in mouse conjunctiva Zhu Hong-Yuan, Riau AK, Beuerman RW. Mol Vis. 2010 ;16:2215-24. 3. A polycationic antimicrobial and biocompatible hydrogel with microbe membrane suctioning Li P, Poon YF, Li W, Zhu Hong-Yuan, Yeap SH, Cao Y, Qi X, Zhou C, Lamrani M, Beuerman RW, Kang ET, Mu Y, Li CM, Chang MW, Leong SS, Chan-Park MB. Nature Material. 2011;10:149-56. 4. Expression of neural receptors in mouse meibomian gland. Zhu Hong-Yuan, Riau AK, Barathi VA, Chew J, Beuerman RW. Cornea. 2010 Jul;29(7):794-801. 5. Stimulation of specific cytokines in human conjunctival epithelial cells by defensins HNP1, HBD2, and HBD3 Li J, Zhu Hong-Yuan, Beuerman RW. Invest Ophthalmol Vis Sci. 2009;50(2):644-53. [...]... sections in hematoxylin for 2-3 minutes (staining time can be modified depending on desired staining density) 4 Wash the slides in running tap water for 2 minutes 5 Identify the nucleus staining with Scott’s tap water for 2 minutes 6 Wash the slides in tap water for 2 minutes 7 Counterstain in eosin for 3 minutes (staining time can be modified depending on desired staining intensity 8 Rinse in 95% ethanol... involvement of specific members of this class of proteins in the development of corneal fibrosis has not been developed 13 1.8.1 Specific aim The specific aims in this study were: 1 To develop a mouse model with corneal fibrosis 2 To dissect specific cytoskeleton regulators active in corneal fibrosis 3 To evaluate the in vivo role of specific cytoskeleton regulators in the regulation of corneal fibrosis. .. cytoskeletal regulators involved in the control of fibrosis in the cornea” 1.8 Hypothesis Based on the strong associations of fibrosis with cytoskeleton regulators , my hypothesis is that this class of proteins which links the intracellular cytoskeleton with the extracellular environment may be a target for therapeutic development for corneal scar; however, as this is a large group of proteins, evidence for involvement... deficient in gelsolin are protected from bleomycininduced fibrosis and gelsolin expression is crucial for the pulmonary fibrosis (Oikonomou N et al 2009) 10 Profilin participates in the reorganization of the cytoskeleton (Hinz B et al 2001b) Profilin mRNA was observed to be up-regulated by using in situ hybridization in hypertrophic fibrosis (Wu J et al 2004) Vinculin, moesin, and ezrin, were elevated in. .. TGF-β1 up-regulated the expression of a variety of cytoskeleton regulators (cofilin, profilin etc.) The details about several cytoskeleton regulators which are implicated to be involved in the fibrosis are shown below Mechanical stimulation is transduced from extracellular environment into intracellular cytoskeleton of myofibroblasts through transmembrane receptorsintegrins that link to intracellular stress... 2003) Cytoskeleton contains three main components: microfilaments, intermediate filaments, and microtubules (Heuser JE et al 1980; Fey EG et al 1984) 1.1.4 Cytoskeleton regulators in biology Rearrangements of cytoskeleton are central to the aforementioned function of cytoskeleton Several studies have highlighted the importance of cytoskeleton regulators in the control of dynamic rearrangements of cytoskeleton. .. fibers through a variety of cytoskeleton regulators (vinculin, talin, paxillin, gelsolin, FAK, ezrin–radixin–moesin proteins) (Geiger B et al 2001) Phenotypic transition of human lung fibroblasts-tomyofibroblasts induced by TGF-β1 depends on integrin signaling via focal adhesion kinase (FAK) (Thannickal VJ et al 2003) FAK, a nonreceptor protein tyrosine kinase, is central for the mechanoresponses, and... rearrangement of cytoskeleton architecture in the corneal stroma keratocytes results in increased expression of α-SMA after rabbit corneal injury Myofibroblast differentiation in mesangial cells is modulated by structure and composition of the cytoskeleton (Patel K et al 2003) Fibroblasts-tomyofibroblasts differentiation in the lung fibrosis is controlled by cytoskeleton 8 tension (Blaauboer ME et al 2011) Cytoskeleton. .. et al 2006) Paxillin has a pivotal role in transducing signals from extracellular environment into intracellular cytoskeleton (Turner CE et al 2000) and mediates the rearrangement of actin cytoskeleton into contractile elements (Cowin AJ et al 2003; O’Kane S et al 1997) The ability of paxillin to modulate fibronectin activities in wound healing may be important in the mechanism of fibrosis- free fetal... consequence of multiple stimuli, but focus is put on TGF-β1 and cytoskeleton regulators in this thesis since the cytoskeletal tension and TGF-β1 play a main role in regulating the acquisition and maintenance of the myofibroblast phenotype (Hinz B et al 2001b; Arora PD et al 1999a; Blaauboer ME et al 2011; Hinz B et al 2003) 1.4.1 TGF-β1 induced fibrosis TGF-β1 is a multifunctional regulatory cytokine and . group of proteins, evidence for involvement of specific members of this class of proteins in the corneal fibrosis has not been developed. Recently, interest in the ERM (ezrin/radixin/moesin). within corneal stroma keratocytes in corneal fibrosis ……………………………………………………………… 47 4.4 Characterization of moesin expression in the corneal fibrosis ……………… 49 4.5 Role of moesin in the corneal. transformation of corneal stroma keratocytes-to-myofibroblasts is predominently responsible for the corneal fibrosis. vii To define the in vivo role of specific cytoskeleton regulators in mediating the

Ngày đăng: 09/09/2015, 18:51

Từ khóa liên quan

Tài liệu cùng người dùng

Tài liệu liên quan