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

Towards topical antifibrotics in tissue engineering and repair

168 313 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

Thông tin cơ bản

Định dạng
Số trang 168
Dung lượng 7,55 MB

Nội dung

Towards Topical Antifibrotics in Tissue Engineering and Repair Chen Zhen Cheng, Clarice (B. Appl. Sci. Hons.) NUS A thesis submitted for the degree of Doctor of Philosophy NUS Graduate School for Integrative Sciences and Engineering National University of Singapore Acknowledgements I would like to thank my supervisor, A/P Michael Raghunath, for infecting me with his passion for research. Working closely with him has taught me the beauty of meticulous work, thorough planning and the importance of having an intellectual sparring partner. I am grateful for Prof. Glenn D. Prestwichs invaluable advice on drug delivery and development of materials. I truly appreciate Dr. Eliana Martinezs help, patience and guidance with the animal studies. Special thanks to my husband and collaborator, Dr. Andrew Krishna Ekaputra, who kept me smiling when times were tough, and generously shared his knowledge and technical skills. My parents are my pillars of support. Endless encouragement from my husband, parents and colleagues in the Tissue Modulation Laboratory gave me strength to get this far. i Table of contents Table of contents . ii Summary . v List of abbreviations vii List of tables . ix List of figures ix Chapter Overview of research project 1.1 Background 1.2 Aims and Objectives 1.3 Research Methodology Chapter Focus on collagen: in vitro systems to study fibrogenesis and antifibrosis — state of the art . 11 2.1 Fibrosis—ubiquitous problem and global burden . 12 2.2 Fibrogenesis in vivo—complexity and key players . 14 2.2.1 Upstream events of fibrosis—cellular players in vivo . 14 2.2.2 “Soluble” factors mediating fibrosis 15 2.3 Understanding the last mile of the fibrotic pathway 16 2.4 Fibrogenesis in vitro—constraints and options . 18 2.4.1 Biosynthetic issues—getting collagen made and deposited in vitro . 19 2.4.2 Quantitative issues—measuring collagen and normalising the data . 22 2.4.3 Qualitative issues—looking beyond collagen I . 25 2.5 Conclusion . 26 2.7 Remarks . 26 Chapter Development of the Scar-in-a-Jar . 28 3.1 Background 29 3.2 Materials and Methods . 35 3.2.1 Fibroblast Cell culture 35 3.2.2 Fibroplasia models 36 3.2.3 Optical analysis . 37 3.2.4 Immunocytochemistry 38 3.2.5 Biochemical analyses- Sodium dodecylsulfate-polyacrylamide gel electrophoresis (SDS-PAGE) . 38 3.2.6 Western blotting . 39 3.2.7 miRNA transfection 39 3.2.8 Statistical Analysis . 39 3.2.9 Materials 40 3.3 Results . 42 3.3.1 Choosing the adequate human cell lines and their properties . 42 3.3.2 Implementation of two collagen deposition protocols 43 3.3.3 Image Processing and Capturing 44 3.3.4 Testing known and novel antifibrotics in comparison with SDS PAGE . 46 3.3.4.1 Epigenetic interference with collagen deposition 46 ii 3.3.4.2 Post-transcriptional interference with collagen deposition . 47 3.3.4.3 Post/Co-translational interference with collagen deposition 49 3.3.4.4 Post-secretional interference with collagen deposition 52 3.5 Conclusion . 65 3.6 Beyond the Scar-in-a-Jar . 66 3.6.1 Materials and Methods 66 3.6.1.1 Procollagen C-proteinase immunoblotting 66 3.6.1.2 FITC-labeling of collagen 67 3.6.1.3 The effect of macromolecular crowding on collagen assembly in vitro . 67 3.6.1.4 The effect of macromolecular crowding on cell-free collagen supramolecular assembly . 68 3.6.2 Results and Discussion . 68 3.6.3 Conclusion . 71 Chapter Peri-implantational fibrosis: a bottleneck in tissue engineering . 72 4.1 Biomaterials in tissue engineering 73 4.2 Foreign body response 73 4.3 Biocompatibility 76 4.4 Vascularization of biomaterials 78 Chapter Development of an antifibrotic and self-vascularizing biomaterial: Design considerations . 82 5.1 Background 83 5.1.1 Prolyl hydroxylase . 83 5.1.1.1 The role of prolyl 4- hydroxylase in collagen biosynthesis 83 5.1.1.2 The role of prolyl hydroxylase in angiogenesis . 83 5.1.2 Prolyl hydroxylase inhibitors 85 5.1.3 Incorporation of PHi into a biomaterial . 89 5.2 Materials and Methods . 93 5.2.1 Fabrication of mPCL/Col-Hep materials 93 5.2.2 Drug release profiles . 94 5.2.3 In vitro cell attachment and infiltration . 94 5.2.4 Bioactivity of incorporated PHi . 96 5.2.5 VEGF reservoir analysis 98 5.2.6 Rat renal pouch model 99 5.2.7 Histological analysis 100 5.2.8 DiI perfusion assay 100 5.2.9 RECA-1 immunohistochemistry and cell scoring . 101 5.2.10 Statistical analysis . 102 5.2.11 Materials 102 5.3 Results . 103 5.3.1 Mode of PHi delivery affects release profile . 103 5.3.2 Incorporation of PHi does not affect ability of mPCL/Col-Hep to support cell attachment and infiltration . 106 5.3.3 PHi released from the biomaterials were bioactive 106 5.3.4 Incorporated PHi induced capillary-like structure formation . 110 5.3.3 De novo synthesised VEGF was stored and released by mPCL/ColHep . 114 5.3.4 Implantation of materials in the rat renal pouch model 116 iii 5.3.5 Peri-implantational fibrosis 119 5.3.6 Functional blood vessel infiltration into implants 120 5.3.7 Endothelial cell infiltration into implants . 121 5.4 Discussion 125 5.4.1 Choice of material(s) and pore size . 126 5.4.2 Drug delivery . 126 5.4.3 Support cell attachment, proliferation and infiltration . 127 5.4.3.1 Antifibrotic property in vitro 128 5.4.3.2 Angiogenic property in vitro . 128 5.4.4 De novo growth factor reservoir . 129 5.4.5 Preliminary in vivo testing 132 5.4.5.1 Efficacy of antifibrotic properties in vivo 133 5.4.5.2 Efficacy of angiogenic properties in vivo . 134 5.5 Conclusion . 135 Chapter Conclusions and Future Work . 136 6.1 Conclusions . 137 6.2 Future work 141 Bibliography . 142 Appendix a A.1 Standard curves for PHi drug release measurement . a A.2 Publications . b A.3 Successful grant applications written/participated in c A.4 Awards . c iv Summary Fibrosis represents a major global disease burden, yet a potent antifibrotic compound is still not in sight. Part of the explanation for this situation is the difficulties that both academic laboratories and R&D departments in the pharmaceutical industry have been facing in re-enacting the fibrotic process in vitro for screening procedures prior to animal testing. Effective in vitro characterization of antifibrotic compounds has been hampered by cell culture settings that are lacking crucial cofactors or are not holistic representations of the biosynthetic and depositional pathway leading to the formation of an insoluble pericellular collagen matrix. Only when collagen has formed a fibrillar matrix that becomes cross-linked, invested with ligands, and can be remodeled and resorbed, the complete picture of fibrogenesis can be reflected in vitro. We developed the Scar-in-a-Jar, which implements for the first time in vitro the complete biosynthetic cascade of collagen matrix formation including complete conversion of procollagen by collagen C-proteinase/BMP-1, its subsequent extracellular deposition and lysyl oxidase-mediated cross-linking. This is achieved by applying the biophysical principle of macromolecular crowding. Collagen matrix deposition velocity and morphology can be controlled using negatively charged crowders in a rapid (2 days) mode and a mixture of neutral crowders in a novel accelerated (6 days) mode. Combined together with quantitative optical bioimaging, this novel system allows for in situ assessment of the area of deposited collagen(s)/cell. A well thought-out in vitro fibrogenesis system represents the missing link between brute force v chemical library screens and rational animal experimentation, thus providing both cost-effectiveness and streamlined procedures towards the development of better antifibrotic drugs. Only upon identification of effective compounds will we then be able to address the current bottleneck in tissue engineering, peri-implantational fibrosis, which not only impede the implants original remedial purpose, but also exacerbates the problem. Compounds from the prolyl-4-hydroxylase inhibitor (PHi) substance class were shown to have antifibrotic potential by the Scar-in-a-Jar, and are further known to stimulate angiogenesis. We believe that local delivery of 2,4-pyridinedicarboxylic acid (PDCA), ciclopirox olamine (CPX) and hydralazine (HDZ) via a material will elicit both antifibrotic and angiogenic tissue behaviour. Besides delivering drugs, some design considerations of such a material include its ability to support cell attachment, proliferation and infiltration, which was attained by our composite material comprising coelectrospun micron-sized medical grade poly(- caprolactone)/collagen (mPCL/Col) with codeposited HeprasilTM, a hyaluronic acid hydrogel containing heparin. mPCL/Col-Hep was used to deliver PHi in two modes, via HeprasilTM (mode 1), or the microfibers (mode 2). Mode was tested in a rat renal pouch model and fibrosis was not apparent around the implanted materials within days, even in controls without PHi. Concentrations of 10 mM PDCA and 10 μM HDZ showed blood vessel infiltration comparable to VEGF delivery, and at this point, served as a proofof-concept that local delivery of PHi can aid angiogenesis. vi List of abbreviations -SMA, alpha smooth muscle actin BAPN, -aminoproprionitrile CLS, capillary-like structure CPX, ciclopirox olamine DiI, 1,1-dioctadecyl-3,3,3-tetramethylindocarbocyanine perchlorate DxS, dextran sulphate (rapid deposition mode) ECM, extracellular matrix ECs, endothelial cells ELISA, enzyme-linked immunosorbent assay EMT, epithelial-mesenchymal transition EVE, excluded-volume effect FBR, foreign body response FBS, fetal bovine serum Fc, ficoll cocktail (accelerated deposition mode) FIT, fluorescence intensity Fn, fibronectin FP, fibroplasia model H&E, hematoxylin and eosin HA, hyaluronic acid HDZ, hydralazine Hep, HeprasilTM HIF-1, hypoxia inducible factor-1 vii HUVECs, human umbilical vein endothelial cells I29c, inhibitor miR29c M29c, mimic miR29c MI, myocardial infarct miRNA, microRNA MMC, macromolecular crowding MMP1, matrix metalloproteinase mPCL, medical grade poly(-caprolactone) OCT, optimal cutting temperature compound PBS, phosphate-buffered saline PCP, procollagen C-proteinase PDCA, 2,4-pyridinedicarboxylic acid PHi, prolyl-4-hydroxylase inhibitors RECA-1, rat endothelial cell antigen-1 SDS-PAGE, SDS-polyacrylamide gel electrophoresis SEM, scanning electron microscopy TGF1, transforming growth factor-1 TSA, trichostatin A VEGF, vascular endothelial growth factor vWF, von Willebrand factor viii List of tables Table Known/novel antifibrotic compounds and their target synthetic keypoints 10 Table IC50 of tested compounds . 41 Table Sumary of antifibrotic effects 59 List of figures Figure Potential points of interference along the collagen biosynthesis pathway. . 18 Figure Accelerated matrix formation under neutral macromolecular crowding at day 2, and analyzed by densitometry and optical analysis 42 Figure Rapid and accelerated collagen deposition modes versus in vitro fibroplasia . 44 Figure Correlation of rapid (DxS) and accelerated (Fc) collagen deposition modes with cell density by the optical analysis method 45 Figure Preservation and quantitation of deposited collagen I and cell enumeration is dependent on the fixative used 47 Figure Image acquisition and quantitation of rapid collagen deposition mode 48 Figure Epigenetic interference of collagen deposition 50 Figure Post-transcriptional regulation of collagen deposition . 51 Figure Optical analyses of the effects of co-translational inhibition of prolyl hydroxylase on collagen I deposition 53 Figure 10 Optical evaluation of extracellular interference with collagen deposition . 55 Figure 11 Optical evaluation of extracellular interference with collagen stabilisation . 56 Figure 12 Optical evaluation of MMP1 activity on deposited collagen I and fibronectin . 57 Figure 13 Biochemical validation of intracellular collagen biosynthesis inhibitors by silver stained SDS PAGE . 58 Figure 14 Biochemical validation of extracellular collagen biosynthesis inhibitors by silver stained SDS PAGE . 59 Figure 15 Enhanced cleavage of the procollagen C-propeptide under macromolecular crowding . 69 Figure 16 Enhanced collagen I supramolecular assembly under macromolecular crowding . 70 Figure 17 Foreign body reaction to and avascular capsule formation around an implanted biomaterial. . 75 Figure 18 Inhibition of prolyl 4-hydroxylase by 2,4-pyridinedicarboxylic acid.84 Figure 19 Regulation of the HIF-1 transcription factor 85 Figure 20 Prolyl hydroxylase inhibitors. 2,4-pyridine dicarboxylic acid is a 2oxoglutarate analogue, and competitively and reversibly binds with prolyl hydroxylase to interfere with its activity. . 86 ix Bibliography 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. Bateman, J.F. and S.B. Golub, Assessment of procollagen processing defects by fibroblasts cultured in the presence of dextran sulphate. Biochem J, 1990. 267(3): p. 573-7. Lareu, R.R., et al., In vitro enhancement of collagen matrix formation and crosslinking for applications in tissue engineering: a preliminary study. Tissue Eng, 2007. 13(2): p. 385-91. Tang, L. and J.W. Eaton, Inflammatory responses to biomaterials. Am J Clin Pathol, 1995. 103(4): p. 466-71. Gristina, A.G., Implant failure and the immuno-incompetent fibro-inflammatory zone. Clin Orthop Relat Res, 1994(298): p. 106-18. Patil, S.D., F. Papadimitrakopoulos, and D.J. Burgess, Dexamethasoneloaded poly(lactic-co-glycolic) acid microspheres/poly(vinyl alcohol) hydrogel composite coatings for inflammation control. Diabetes Technol Ther, 2004. 6(6): p. 887-97. Chen, C.Z. and M. Raghunath, Focus on collagen: in vitro systems to study fibrogenesis and antifibrosis -- state of the art. Fibrogenesis Tissue Repair, 2009. 2(1): p. 7. Ratner, B.D. and S.J. Bryant, Biomaterials: where we have been and where we are going. Annu Rev Biomed Eng, 2004. 6: p. 41-75. Corpechot, C., et al., Hypoxia-induced VEGF and collagen I expressions are associated with angiogenesis and fibrogenesis in experimental cirrhosis. Hepatology, 2002. 35(5): p. 1010-21. Kannan, R.Y., et al., The roles of tissue engineering and vascularisation in the development of micro-vascular networks: a review. Biomaterials, 2005. 26(14): p. 1857-75. Chen, C.Z., et al., The Scar-in-a-Jar: studying potential antifibrotic compounds from the epigenetic to extracellular level in a single well. Br J Pharmacol, 2009. 158(5): p. 1196-209. Raghunath, M., et al., Pharmacologically induced angiogenesis in transgenic zebrafish. Biochem Biophys Res Commun, 2009. 378(4): p. 766-71. Clark, R.A., et al., TGF-beta stimulates cultured human fibroblasts to proliferate and produce tissue-like fibroplasia: a fibronectin matrix-dependent event. J Cell Physiol, 1997. 170(1): p. 69-80. Fish, P.V., et al., Potent and selective nonpeptidic inhibitors of procollagen Cproteinase. J Med Chem, 2007. 50(15): p. 3442-56. Miranda, K.C., et al., A pattern-based method for the identification of MicroRNA binding sites and their corresponding heteroduplexes. Cell, 2006. 126(6): p. 1203-17. Meltzer, E.B. and P.W. Noble, Idiopathic pulmonary fibrosis. Orphanet J Rare Dis, 2008. 3: p. 8. WHO, The Global Burden of Disease 2004 update. 2004, Geneva, Switzerland: WHO press. Cleutjens, J.P., et al., Regulation of collagen degradation in the rat myocardium after infarction. J Mol Cell Cardiol, 1995. 27(6): p. 1281-92. Schmermund, A., et al., Coronary atherosclerosis in unheralded sudden coronary death under age 50: histo-pathologic comparison with 'healthy' subjects dying out of hospital. Atherosclerosis, 2001. 155(2): p. 499-508. Hilborn, J. and L.M. Bjursten, A new and evolving paradigm for biocompatibility. J Tissue Eng Regen Med, 2007. 1(2): p. 110-9. 142 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. 37. 38. Thevenot, P., W. Hu, and L. Tang, Surface chemistry influences implant biocompatibility. Curr Top Med Chem, 2008. 8(4): p. 270-80. Schiavone Panni, A., et al., The bone-ligament junction: a comparison between biological and artificial ACL reconstruction. Knee Surg Sports Traumatol Arthrosc, 1993. 1(1): p. 9-12. Prescott, R.J., W.G. Ryan, and D.L. Bisset, Histopathological features of failed prosthetic Leeds-Keio anterior cruciate ligaments. J Clin Pathol, 1994. 47(4): p. 375-6. Koschwanez, H.E., et al., In vitro and in vivo characterization of porous polyL-lactic acid coatings for subcutaneously implanted glucose sensors. J Biomed Mater Res A, 2008. 87(3): p. 792-807. Klueh, U., D.I. Dorsky, and D.L. Kreutzer, Enhancement of implantable glucose sensor function in vivo using gene transfer-induced neovascularization. Biomaterials, 2005. 26(10): p. 1155-63. Jerosch, J. and A.M. Aldawoudy, Arthroscopic treatment of patients with moderate arthrofibrosis after total knee replacement. Knee Surg Sports Traumatol Arthrosc, 2007. 15(1): p. 71-7. Ries, M.D. and M. Badalamente, Arthrofibrosis after total knee arthroplasty. Clin Orthop Relat Res, 2000(380): p. 177-83. Ulrich, D., et al., Effect of tissue inhibitors of metalloproteinases and matrix metalloproteinases on capsular formation around smooth and textured silicone gel implants. Aesthetic Plast Surg, 2009. 33(4): p. 555-62. Spano, A., et al., Reduction of capsular thickness around silicone breast implants by zafirlukast in rats. Eur Surg Res, 2008. 41(1): p. 8-14. Rafael, E., et al., Improved survival of macroencapsulated islets of Langerhans by preimplantation of the immunoisolating device: a morphometric study. Cell Transplant, 2003. 12(4): p. 407-12. Dufrane, D., et al., Six-month survival of microencapsulated pig islets and alginate biocompatibility in primates: proof of concept. Transplantation, 2006. 81(9): p. 1345-53. Bellucci, S., et al., Multiwalled carbon nanotube buckypaper: toxicology and biological effects in vitro and in vivo. Nanomed, 2009. 4(5): p. 531-40. Sahiner, N., et al., Creation of a drug-coated glaucoma drainage device using polymer technology: in vitro and in vivo studies. Arch Ophthalmol, 2009. 127(4): p. 448-53. Mavrikakis, I., et al., Medpor lower eyelid spacer: does it biointegrate? Orbit, 2009. 28(1): p. 58-62. Tang, L. and W. Hu, Molecular determinants of biocompatibility. Expert Rev. Med. Devices, 2005. 2(4): p. 493-500. Riccetto, C., et al., Impact of the structure of polypropylene meshes in local tissue reaction: in vivo stereological study. Int Urogynecol J Pelvic Floor Dysfunct, 2008. 19(8): p. 1117-23. Liu, L., et al., Reduced foreign body reaction to implanted biomaterials by surface treatment with oriented osteopontin. J Biomater Sci Polym Ed, 2008. 19(6): p. 821-35. Yoon, S.J., et al., Reduction of inflammatory reaction of poly(d,l-lactic-coglycolic Acid) using demineralized bone particles. Tissue Eng Part A, 2008. 14(4): p. 539-47. Chen, Z.C., et al., In vitro and in vivo analysis of co-electrospun scaffolds made of medical grade poly(epsilon-caprolactone) and porcine collagen. J Biomater Sci Polym Ed, 2008. 19(5): p. 693-707. 143 39. 40. 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. 57. 58. 59. Mendes, J.B., et al., Cilostazol and pentoxifylline decrease angiogenesis, inflammation, and fibrosis in sponge-induced intraperitoneal adhesion in mice. Life Sci, 2009. 84(15-16): p. 537-43. Wynn, T.A., Common and unique mechanisms regulate fibrosis in various fibroproliferative diseases. J Clin Invest, 2007. 117(3): p. 524-9. Li, C., et al., Homing of bone marrow mesenchymal stem cells mediated by sphingosine 1-phosphate contributes to liver fibrosis. J Hepatol, 2009. 50(6): p. 1174-83. Zavadil, J. and E.P. Bottinger, TGF-beta and epithelial-to-mesenchymal transitions. Oncogene, 2005. 24(37): p. 5764-74. Guarino, M., A. Tosoni, and M. Nebuloni, Direct contribution of epithelium to organ fibrosis: epithelial-mesenchymal transition. Hum Pathol, 2009. Raghunath, M., et al., The cutaneous microfibrillar apparatus contains latent transforming growth factor-beta binding protein-1 (LTBP-1) and is a repository for latent TGF-beta1. J Invest Dermatol, 1998. 111(4): p. 559-64. Droguett, R., et al., Extracellular proteoglycans modify TGF-beta bioavailability attenuating its signaling during skeletal muscle differentiation. Matrix Biol, 2006. 25(6): p. 332-41. Zhang, Z., et al., Recombinant human decorin inhibits cell proliferation and downregulates TGF-beta1 production in hypertrophic scar fibroblasts. Burns, 2007. 33(5): p. 634-41. O'Kane, S. and M.W. Ferguson, Transforming growth factor beta s and wound healing. Int J Biochem Cell Biol, 1997. 29(1): p. 63-78. Ponticos, M., et al., Pivotal role of connective tissue growth factor in lung fibrosis: MAPK-dependent transcriptional activation of type I collagen. Arthritis Rheum, 2009. 60(7): p. 2142-55. Liu, Y., et al., Therapeutic targeting of the PDGF and TGF-beta-signaling pathways in hepatic stellate cells by PTK787/ZK22258. Lab Invest, 2009. Wynn, T., Cellular and molecular mechanisms of fibrosis. J Pathol, 2007. 214(2): p. 199-210. Wynn, T.A., et al., An IL-12-based vaccination method for preventing fibrosis induced by schistosome infection. Nature, 1995. 376(6541): p. 594-6. Wynn, T.A., Fibrotic disease and the T(H)1/T(H)2 paradigm. Nat Rev Immunol, 2004. 4(8): p. 583-94. Grutter, C., et al., A cytokine-neutralizing antibody as a structural mimetic of receptor interactions. Proc Natl Acad Sci U S A, 2008. 105(51): p. 20251-6. Sumioka, T., et al., Inhibitory effect of blocking TGF-beta/Smad signal on injury-induced fibrosis of corneal endothelium. Mol Vis, 2008. 14: p. 2272-81. Caballero, S., et al., Anti-sphingosine-1-phosphate monoclonal antibodies inhibit angiogenesis and sub-retinal fibrosis in a murine model of laserinduced choroidal neovascularization. Exp Eye Res, 2009. 88(3): p. 367-77. Ikawa, Y., et al., Neutralizing monoclonal antibody to human connective tissue growth factor ameliorates transforming growth factor-beta-induced mouse fibrosis. J Cell Physiol, 2008. 216(3): p. 680-7. Lin, X., et al., Controlled release of matrix metalloproteinase with or without skeletal myoblasts transplantation improves cardiac function of rat hearts with chronic myocardial infarction. Tissue Eng Part A, 2009. 15(9): p. 2699-706. Kaar, J.L., et al., Matrix metalloproteinase-1 treatment of muscle fibrosis. Acta Biomater, 2008. 4(5): p. 1411-20. Hu, P.F., et al., Adenovirus-mediated transfer of siRNA against PAI-1 mRNA ameliorates hepatic fibrosis in rats. J Hepatol, 2009. 51(1): p. 102-13. 144 60. 61. 62. 63. 64. 65. 66. 67. 68. 69. 70. 71. 72. 73. 74. 75. 76. Sato, Y., et al., Resolution of liver cirrhosis using vitamin A-coupled liposomes to deliver siRNA against a collagen-specific chaperone. Nat Biotechnol, 2008. 26(4): p. 431-42. Ferguson, M.W., et al., Prophylactic administration of avotermin for improvement of skin scarring: three double-blind, placebo-controlled, phase I/II studies. Lancet, 2009. 373(9671): p. 1264-74. Wang, Z., et al., SAHA- a potential epigenetic therapeutic agent for lung fibrosis? Eur Respir J, 2009. Ghosh, A.K., et al., Trichostatin A blocks TGF-beta-induced collagen gene expression in skin fibroblasts: Involvement of Sp1. Biochemical and Biophysical Research Communications, 2007. 354(2): p. 420-426. Zion, O., et al., Inhibition of transforming growth factor beta signaling by halofuginone as a modality for pancreas fibrosis prevention. Pancreas, 2009. 38(4): p. 427-35. Olbrich, K.C., et al., Halofuginone inhibits collagen deposition in fibrous capsules around implants. Ann Plast Surg, 2005. 54(3): p. 293-6;discussion 296. Cheng, K., N. Yang, and R.I. Mahato, TGF-beta1 gene silencing for treating liver fibrosis. Mol Pharm, 2009. 6(3): p. 772-9. van Rooij, E., et al., Dysregulation of microRNAs after myocardial infarction reveals a role of miR-29 in cardiac fibrosis. Proc Natl Acad Sci U S A, 2008. 105(35): p. 13027-32. Chen, C., et al., The Scar-in-a-Jar: Studying antifibrotic lead compounds from the epigenetic to extracellular level in a single well. Br J Pharmacol, 2009: p. In press. Tschank, G., et al., Inhibition of prolyl hydroxylation and procollagen processing in chick-embryo calvaria by a derivative of pyridine-2,4dicarboxylate. Characterization of the diethyl ester as a proinhibitor. Biochem J, 1991. 275 ( Pt 2): p. 469-76. Lareu, R.R., et al., Collagen matrix deposition is dramatically enhanced in vitro when crowded with charged macromolecules: the biological relevance of the excluded volume effect. FEBS Lett, 2007. 581(14): p. 2709-14. Sauk, J.J., N. Nikitakis, and H. Siavash, Hsp47 a novel collagen binding serpin chaperone, autoantigen and therapeutic target. Front Biosci, 2005. 10: p. 107-18. Tambara, K., et al., Administration of control-released hepatocyte growth factor enhances the efficacy of skeletal myoblast transplantation in rat infarcted hearts by greatly increasing both quantity and quality of the graft. Circulation, 2005. 112(9 Suppl): p. I129-34. Mias, C., et al., Mesenchymal Stem Cells Promote Matrix Metalloproteinase Secretion By Cardiac Fibroblasts And Reduce Cardiac Ventricular Fibrosis After Myocardial Infarction. Stem Cells, 2009. Xu, Q., et al., In vitro models of TGF-beta-induced fibrosis suitable for highthroughput screening of antifibrotic agents. Am J Physiol Renal Physiol, 2007. 293(2): p. F631-40. Hata, R. and H. Senoo, L-ascorbic acid 2-phosphate stimulates collagen accumulation, cell proliferation, and formation of a three-dimensional tissuelike substance by skin fibroblasts. J Cell Physiol, 1989. 138(1): p. 8-16. Murad, S., et al., Collagen synthesis in cultured human skin fibroblasts: effect of ascorbic acid and its analogs. J Invest Dermatol, 1983. 81(2): p. 158-62. 145 77. 78. 79. 80. 81. 82. 83. 84. 85. 86. 87. 88. 89. 90. 91. 92. 93. 94. 95. Yu, Y.M., et al., Arginine and ornithine kinetics in severely burned patients: increased rate of arginine disposal. Am J Physiol Endocrinol Metab, 2001. 280(3): p. E509-17. Yu, Y.M., et al., Relations among arginine, citrulline, ornithine, and leucine kinetics in adult burn patients. Am J Clin Nutr, 1995. 62(5): p. 960-8. Barbul, A., Proline precursors to sustain Mammalian collagen synthesis. J Nutr, 2008. 138(10): p. 2021S-2024S. Ng, K.W., et al., Amino acids supply in culture media is not a limiting factor in the matrix synthesis of engineered cartilage tissue. Amino Acids, 2008. 35(2): p. 433-8. Peng, Y. and M. Raghunath, Learning from Nature: emulating macromolecular crowding to drive extracellular matrix enhancement for the creation of connective tissue, in Tissue Engineering. 2009, In Tech: Vienna. p. In press. Woessner, J.F., Jr., The determination of hydroxyproline in tissue and protein samples containing small proportions of this imino acid. Arch Biochem Biophys, 1961. 93: p. 440-7. Sweat, F., H. Puchtler, and S.I. Rosenthal, Sirius Red F3ba as a Stain for Connective Tissue. Arch Pathol, 1964. 78: p. 69-72. Junqueira, L.C., G. Bignolas, and R.R. Brentani, Picrosirius staining plus polarization microscopy, a specific method for collagen detection in tissue sections. Histochem J, 1979. 11(4): p. 447-55. Walsh, B.J., et al., Microplate reader-based quantitation of collagens. Anal Biochem, 1992. 203(2): p. 187-90. Houghton, P.E., et al., A simple method to assess the relative amount of collagen deposition in wounded fetal mouse limbs. Wound Repair Regen, 1996. 4(4): p. 489-95. Lareu, R.R., et al., Essential modification of the Sircol Collagen Assay for the accurate quantification of collagen content in complex protein solutions. Acta Biomater, 2010. 6(8): p. 3146-51. Tullberg-Reinert, H. and G. Jundt, In situ measurement of collagen synthesis by human bone cells with a Sirius Red-based colorimetric microassay: effects of transforming growth factor beta and ascorbic acid 2-phosphate. Histochemistry and Cell Biology, 1999. 112(4): p. 271-276. Campa, J.S., R.J. McAnulty, and G.J. Laurent, Application of high-pressure liquid chromatography to studies of collagen production by isolated cells in culture. Anal Biochem, 1990. 186(2): p. 257-63. Stimler, N.P., High-performance liquid chromatographic quantitation of collagen biosynthesis in explant cultures. Anal Biochem, 1984. 142(1): p. 1038. Tredget, E.E., et al., Determination of 4-hydroxyproline in collagen by gas chromatography/mass spectrometry. Anal Biochem, 1990. 190(2): p. 259-65. Limeback, H.F. and J. Sodek, Procollagen synthesis and processing in periodontal ligament in vivo and in vitro. A comparative study using slab-gel fluorography. Eur J Biochem, 1979. 100(2): p. 541-50. Raghunath, M., et al., Prenatal diagnosis of collagen disorders by direct biochemical analysis of chorionic villus biopsies. Pediatr Res, 1994. 36(4): p. 441-8. Ramshaw, J.A. and J.A. Werkmeister, Electrophoresis and electroblotting of native collagens. Anal Biochem, 1988. 168(1): p. 82-7. Kirchhofer, D., C.A. Reinhardt, and G. Zbinden, Collagen synthesis in growing human skin fibroblasts. Exp Cell Biol, 1986. 54(4): p. 177-82. 146 96. 97. 98. 99. 100. 101. 102. 103. 104. 105. 106. 107. 108. 109. 110. 111. 112. 113. 114. Acharya, P.S., et al., Fibroblast activation protein: a serine protease expressed at the remodeling interface in idiopathic pulmonary fibrosis. Hum Pathol, 2006. 37(3): p. 352-60. Wynn, T.A. and L. Barron, Macrophages: master regulators of inflammation and fibrosis. Semin Liver Dis, 2010. 30(3): p. 245-57. Paternostro, C., et al., Hypoxia, angiogenesis and liver fibrogenesis in the progression of chronic liver diseases. World J Gastroenterol, 2010. 16(3): p. 281-8. Terashima, H., et al., A sensitive short-term evaluation of antifibrotic effects using newly established type I collagen reporter transgenic rats. Am J Physiol Renal Physiol, 2010. 299(4): p. F792-801. Anderson, S.M. and R.J. Elliott, Evaluation of a new, rapid collagen assay. Biochem Soc Trans, 1991. 19(4): p. 389S. Antonini, J.M., et al., Application of laser scanning confocal microscopy in the analysis of particle-induced pulmonary fibrosis. Toxicological Sciences, 1999. 51(1): p. 126-134. Fulton, A.B., How crowded is the cytoplasm? Cell, 1982. 30(2): p. 345-7. Minton, A.P., Confinement as a determinant of macromolecular structure and reactivity. II. Effects of weakly attractive interactions between confined macrosolutes and confining structures. Biophys J, 1995. 68(4): p. 1311-22. Ellis, R.J. and A.P. Minton, Cell biology: join the crowd. Nature, 2003. 425(6953): p. 27-8. Ebel, C. and G. Zaccai, Crowding in extremophiles: linkage between solvation and weak protein-protein interactions, stability and dynamics, provides insight into molecular adaptation. J Mol Recognit, 2004. 17(5): p. 382-9. Ellis, R.J., Macromolecular crowding: obvious but underappreciated. Trends Biochem Sci, 2001. 26(10): p. 597-604. Zimmerman, S.B. and A.P. Minton, Macromolecular crowding: biochemical, biophysical, and physiological consequences. Annu Rev Biophys Biomol Struct, 1993. 22: p. 27-65. Hall, D. and A.P. Minton, Macromolecular crowding: qualitative and semiquantitative successes, quantitative challenges. Biochim Biophys Acta, 2003. 1649(2): p. 127-39. Cheung, M.S., D. Klimov, and D. Thirumalai, Molecular crowding enhances native state stability and refolding rates of globular proteins. Proc Natl Acad Sci U S A, 2005. 102(13): p. 4753-8. Goobes, R., et al., Metabolic buffering exerted by macromolecular crowding on DNA-DNA interactions: origin and physiological significance. Biochemistry, 2003. 42(8): p. 2431-40. Minton, A.P., Models for excluded volume interaction between an unfolded protein and rigid macromolecular cosolutes: macromolecular crowding and protein stability revisited. Biophys J, 2005. 88(2): p. 971-85. Ellis, R.J., Macromolecular crowding: an important but neglected aspect of the intracellular environment. Curr Opin Struct Biol, 2001. 11(1): p. 114-9. Minton, A.P., The influence of macromolecular crowding and macromolecular confinement on biochemical reactions in physiological media. J Biol Chem, 2001. 276(14): p. 10577-80. Chen, C., et al., Applying macromolecular crowding to enhance extracellular matrix deposition and its remodelling in vitro for tissue engineering and cellbased therapies. Advanced Drug Delivery Reviews, In press. 147 115. 116. 117. 118. 119. 120. 121. 122. 123. 124. 125. 126. 127. 128. 129. 130. 131. 132. 133. Furumai, R., et al., Potent histone deacetylase inhibitors built from trichostatin A and cyclic tetrapeptide antibiotics including trapoxin. Proc Natl Acad Sci U S A, 2001. 98(1): p. 87-92. Tschank, G., et al., Pyridinedicarboxylates, the first mechanism-derived inhibitors for prolyl 4-hydroxylase, selectively suppress cellular hydroxyprolyl biosynthesis. Decrease in interstitial collagen and Clq secretion in cell culture. Biochem J, 1987. 248(3): p. 625-33. Clement, P.M., et al., The antifungal drug ciclopirox inhibits deoxyhypusine and proline hydroxylation, endothelial cell growth and angiogenesis in vitro. Int J Cancer, 2002. 100(4): p. 491-8. Burchardt, E.-R., et al., Phosphinate peptide analogs for the treatment of fibrotic disorders, USPTO, Editor. 2004: USA. Wilmarth, K.R. and J.R. Froines, In vitro and in vivo inhibition of lysyl oxidase by aminopropionitriles. J Toxicol Environ Health, 1992. 37(3): p. 411-23. Huber, L.C., et al., Trichostatin a prevents the accumulation of extracellular matrix in a mouse model of bleomycin-induced skin fibrosis. Arthritis and Rheumatism, 2007. 56(8): p. 2755-2764. Gross, J. and Y. Nagai, Specific degradation of the collagen molecule by tadpole collagenolytic enzyme. Proc Natl Acad Sci U S A, 1965. 54(4): p. 1197-204. Varga, J. and S.A. Jimenez, Stimulation of normal human fibroblast collagen production and processing by transforming growth factor-beta. Biochem Biophys Res Commun, 1986. 138(2): p. 974-80. Overall, C.M., J.L. Wrana, and J. Sodek, Independent regulation of collagenase, 72-kDa progelatinase, and metalloendoproteinase inhibitor expression in human fibroblasts by transforming growth factor-beta. J Biol Chem, 1989. 264(3): p. 1860-9. Kisseleva, T. and D.A. Brenner, Mechanisms of fibrogenesis. Exp Biol Med (Maywood), 2008. 233(2): p. 109-22. Wetzel, M., et al., Effect of trichostatin A, a histone deacetylase inhibitor, on glioma proliferation in vitro by inducing cell cycle arrest and apoptosis. J Neurosurg, 2005. 103(6 Suppl): p. 549-56. Platta, C.S., et al., The HDAC inhibitor trichostatin A inhibits growth of small cell lung cancer cells. J Surg Res, 2007. 142(2): p. 219-26. Rombouts, K., et al., Trichostatin A, a histone deacetylase inhibitor, suppresses collagen synthesis and prevents TGF-beta(1)-induced fibrogenesis in skin fibroblasts. Experimental Cell Research, 2002. 278(2): p. 184-197. Wang, Z., et al., Suberoylanilide hydroxamic acid: a potential epigenetic therapeutic agent for lung fibrosis? Eur Respir J, 2009. 34(1): p. 145-55. Asikainen, T.M., et al., Stimulation of HIF-1alpha, HIF-2alpha, and VEGF by prolyl 4-hydroxylase inhibition in human lung endothelial and epithelial cells. Free Radic Biol Med, 2005. 38(8): p. 1002-13. Burchardt, E.-R., et al., Phosphinate peptide analogs for the treatment of fibrotic disorders. 1999: US. Robinson, L.A., et al., Novel inhibitors of procollagen C-proteinase. Part 2: glutamic acid hydroxamates. Bioorg Med Chem Lett, 2003. 13(14): p. 2381-4. Pinnell, S.R., Molecular defects in the Ehlers-Danlos syndrome. J Invest Dermatol, 1982. 79 Suppl 1: p. 90s-92s. Kagan, H.M., Intra- and extracellular enzymes of collagen biosynthesis as biological and chemical targets in the control of fibrosis. Acta Trop, 2000. 77(1): p. 147-52. 148 134. 135. 136. 137. 138. 139. 140. 141. 142. 143. 144. 145. 146. 147. 148. 149. 150. 151. 152. 153. 154. Vater, C.A., E.D. Harris, Jr., and R.C. Siegel, Native cross-links in collagen fibrils induce resistance to human synovial collagenase. Biochem J, 1979. 181(3): p. 639-45. Layman, D.L., E.B. McGoodwin, and G.R. Martin, The nature of the collagen synthesized by cultured human fibroblasts. Proc Natl Acad Sci U S A, 1971. 68(2): p. 454-8. Lombard, C., J. Saulnier, and J. Wallach, Assays of matrix metalloproteinases (MMPs) activities: a review. Biochimie, 2005. 87(3-4): p. 265-72. Troeberg, L. and H. Nagase, Analysis of TIMP expression and activity. Methods Mol Med, 2007. 135: p. 251-67. Nakai, A., et al., Involvement of the stress protein HSP47 in procollagen processing in the endoplasmic reticulum. J Cell Biol, 1992. 117(4): p. 903-14. Lewis, B.P., et al., Prediction of mammalian microRNA targets. Cell, 2003. 115(7): p. 787-98. Overall, C.M., A microtechnique for dialysis of small volume solutions with quantitative recoveries. Anal Biochem, 1987. 165(1): p. 208-14. Jones, K.S., Effects of biomaterial-induced inflammation on fibrosis and rejection. Semin Immunol, 2008. 20(2): p. 130-6. Anderson, J.M., A. Rodriguez, and D.T. Chang, Foreign body reaction to biomaterials. Semin Immunol, 2008. 20(2): p. 86-100. Ratner, B.D., Reducing capsular thickness and enhancing angiogenesis around implant drug release systems. J Control Release, 2002. 78(1-3): p. 211-8. Johnston, R.B., Jr., Current concepts: immunology. Monocytes and macrophages. N Engl J Med, 1988. 318(12): p. 747-52. Pober, J.S. and R.S. Cotran, The role of endothelial cells in inflammation. Transplantation, 1990. 50(4): p. 537-44. Pierce, G.F., et al., Role of platelet-derived growth factor in wound healing. J Cell Biochem, 1991. 45(4): p. 319-26. Labat-Robert, J., M. Bihari-Varga, and L. Robert, Extracellular matrix. FEBS Lett, 1990. 268(2): p. 386-93. Morais, J.M., F. Papadimitrakopoulos, and D.J. Burgess, Biomaterials/tissue interactions: possible solutions to overcome foreign body response. Aaps J, 2010. 12(2): p. 188-96. Cejas, M.A., et al., Thrombogenic collagen-mimetic peptides: Self-assembly of triple helix-based fibrils driven by hydrophobic interactions. Proc Natl Acad Sci U S A, 2008. 105(25): p. 8513-8. Schoen, F.J. and J.M. Anderson, Host response to biomaterials and their evaluation., in Biomaterials science: an introduction to materials in medicine, B.D. Ratner, F.J. Schoen, and J.E. Lemons, Editors. 2004, Elsevier: San Diego. p. 293-6. Power, K.A., K.T. Fitzgerald, and W.M. Gallagher, Examination of cell-hostbiomaterial interactions via high-throughput technologies: A re-appraisal. Biomaterials, 2010. 31(26): p. 6667-6674. Le, S.J., et al., Gene expression profile of the fibrotic response in the peritoneal cavity. Differentiation, 2010. 79(4-5): p. 232-43. Ulici, V., et al., Regulation of gene expression by PI3K in mouse growth plate chondrocytes. PLoS One, 2010. 5(1): p. e8866. Minogue, B.M., et al., Transcriptional profiling of bovine intervertebral disc cells: implications for identification of normal and degenerate human intervertebral disc cell phenotypes. Arthritis Res Ther, 2010. 12(1): p. R22. 149 155. 156. 157. 158. 159. 160. 161. 162. 163. 164. 165. 166. 167. 168. 169. 170. 171. 172. 173. Dinnes, D.L., et al., Material surfaces affect the protein expression patterns of human macrophages: A proteomics approach. J Biomed Mater Res A, 2007. 80(4): p. 895-908. Zuckerman, S.T., J.F. Brown, and W.J. Kao, Identification of regulatory Hck and PAI-2 proteins in the monocyte response to PEG-containing matrices. Biomaterials, 2009. 30(23-24): p. 3825-33. Hickey, T., et al., In vivo evaluation of a dexamethasone/PLGA microsphere system designed to suppress the inflammatory tissue response to implantable medical devices. J Biomed Mater Res, 2002. 61(2): p. 180-7. Wang, C., et al., Synthesis and performance of novel hydrogels coatings for implantable glucose sensors. Biomacromolecules, 2008. 9(2): p. 561-7. Wood, R.C., E.L. LeCluyse, and J.A. Fix, Assessment of a model for measuring drug diffusion through implant-generated fibrous capsule membranes. Biomaterials, 1995. 16(12): p. 957-9. Nishi, O., et al., Intercapsular cataract surgery with lens epithelial cell removal. Part IV: Capsular fibrosis induced by poly(methyl methacrylate). J Cataract Refract Surg, 1991. 17(4): p. 471-7. Yu, B., et al., Use of hydrogel coating to improve the performance of implanted glucose sensors. Biosens Bioelectron, 2008. 23(8): p. 1278-84. Scherer, S.S., et al., Poly-N-acetyl glucosamine nanofibers: a new bioactive material to enhance diabetic wound healing by cell migration and angiogenesis. Ann Surg, 2009. 250(2): p. 322-30. Ravindran, J., et al., Thymoquinone poly (lactide-co-glycolide) nanoparticles exhibit enhanced anti-proliferative, anti-inflammatory, and chemosensitization potential. Biochem Pharmacol, 2010. 79(11): p. 1640-7. Chow, E.K., et al., Copolymeric nanofilm platform for controlled and localized therapeutic delivery. ACS Nano, 2008. 2(1): p. 33-40. Folkman, J. and P.A. D'Amore, Blood vessel formation: what is its molecular basis? Cell, 1996. 87(7): p. 1153-5. Jain, R.K., Molecular regulation of vessel maturation. Nat Med, 2003. 9(6): p. 685-93. Bell, S.E., et al., Differential gene expression during capillary morphogenesis in 3D collagen matrices: regulated expression of genes involved in basement membrane matrix assembly, cell cycle progression, cellular differentiation and G-protein signaling. J Cell Sci, 2001. 114(Pt 15): p. 2755-73. Davis, G.E. and D.R. Senger, Endothelial extracellular matrix: biosynthesis, remodeling, and functions during vascular morphogenesis and neovessel stabilization. Circ Res, 2005. 97(11): p. 1093-107. Moon, J.J., et al., Biomimetic hydrogels with pro-angiogenic properties. Biomaterials, 2010. Golub, J.S., et al., Sustained VEGF delivery via PLGA nanoparticles promotes vascular growth. Am J Physiol Heart Circ Physiol, 2010. 298(6): p. H1959-65. Elia, R., et al., Stimulation of in vivo angiogenesis by in situ crosslinked, dual growth factor-loaded, glycosaminoglycan hydrogels. Biomaterials, 2010. 31(17): p. 4630-8. Chiu, L.L. and M. Radisic, Scaffolds with covalently immobilized VEGF and Angiopoietin-1 for vascularization of engineered tissues. Biomaterials, 2010. 31(2): p. 226-41. Gelain, F., L.D. Unsworth, and S. Zhang, Slow and sustained release of active cytokines from self-assembling peptide scaffolds. J Control Release, 2010. 145(3): p. 231-9. 150 174. 175. 176. 177. 178. 179. 180. 181. 182. 183. 184. 185. 186. 187. 188. 189. 190. 191. 192. 193. Yang, F., et al., Genetic engineering of human stem cells for enhanced angiogenesis using biodegradable polymeric nanoparticles. Proc Natl Acad Sci U S A, 2010. 107(8): p. 3317-22. Simons, M. and J.A. Ware, Therapeutic angiogenesis in cardiovascular disease. Nat Rev Drug Discov, 2003. 2(11): p. 863-71. Nagy, J.A., et al., Vascular permeability factor/vascular endothelial growth factor induces lymphangiogenesis as well as angiogenesis. J Exp Med, 2002. 196(11): p. 1497-506. Henry, T.D., et al., The VIVA trial: Vascular endothelial growth factor in Ischemia for Vascular Angiogenesis. Circulation, 2003. 107(10): p. 1359-65. Freedman, S.B. and J.M. Isner, Therapeutic angiogenesis for coronary artery disease. Ann Intern Med, 2002. 136(1): p. 54-71. Faure, C., et al., Functionalization of matrices by cyclically stretched osteoblasts through matrix targeting of VEGF. Biomaterials, 2010. 31(25): p. 6477-84. Chen, L., et al., Loading of VEGF to the heparin cross-linked demineralized bone matrix improves vascularization of the scaffold. J Mater Sci Mater Med, 2010. 21(1): p. 309-17. Cabric, S., et al., Anchoring of vascular endothelial growth factor to surfaceimmobilized heparin on pancreatic islets: implications for stimulating islet angiogenesis. Tissue Eng Part A, 2010. 16(3): p. 961-70. Grieb, G., et al., Tissue substitutes with improved angiogenic capabilities: an in vitro investigation with endothelial cells and endothelial progenitor cells. Cells Tissues Organs, 2010. 191(2): p. 96-104. Poh, C.K., et al., The effect of VEGF functionalization of titanium on endothelial cells in vitro. Biomaterials, 2010. 31(7): p. 1578-85. Ozawa, C.R., et al., Microenvironmental VEGF concentration, not total dose, determines a threshold between normal and aberrant angiogenesis. J Clin Invest, 2004. 113(4): p. 516-27. Silva, E.A. and D.J. Mooney, Effects of VEGF temporal and spatial presentation on angiogenesis. Biomaterials, 2010. 31(6): p. 1235-41. Sasisekharan, R. and G. Venkataraman, Heparin and heparan sulfate: biosynthesis, structure and function. Curr Opin Chem Biol, 2000. 4(6): p. 62631. Stringer, S.E. and J.T. Gallagher, Heparan sulphate. Int J Biochem Cell Biol, 1997. 29(5): p. 709-14. Klagsbrun, M. and A. Baird, A dual receptor system is required for basic fibroblast growth factor activity. Cell, 1991. 67(2): p. 229-31. Yayon, A., et al., Cell surface, heparin-like molecules are required for binding of basic fibroblast growth factor to its high affinity receptor. Cell, 1991. 64(4): p. 841-8. Macri, L., D. Silverstein, and R.A. Clark, Growth factor binding to the pericellular matrix and its importance in tissue engineering. Adv Drug Deliv Rev, 2007. 59(13): p. 1366-81. Ruhrberg, C., Growing and shaping the vascular tree: multiple roles for VEGF. Bioessays, 2003. 25(11): p. 1052-60. Carmeliet, P., et al., Impaired myocardial angiogenesis and ischemic cardiomyopathy in mice lacking the vascular endothelial growth factor isoforms VEGF164 and VEGF188. Nat Med, 1999. 5(5): p. 495-502. Tessler, S., et al., Heparin modulates the interaction of VEGF165 with soluble and cell associated flk-1 receptors. J Biol Chem, 1994. 269(17): p. 12456-61. 151 194. 195. 196. 197. 198. 199. 200. 201. 202. 203. 204. 205. 206. 207. 208. 209. 210. 211. Kendall, R.L. and K.A. Thomas, Inhibition of vascular endothelial cell growth factor activity by an endogenously encoded soluble receptor. Proc Natl Acad Sci U S A, 1993. 90(22): p. 10705-9. Rolny, C., et al., Heparin amplifies platelet-derived growth factor (PDGF)- BBinduced PDGF alpha -receptor but not PDGF beta -receptor tyrosine phosphorylation in heparan sulfate-deficient cells. Effects on signal transduction and biological responses. J Biol Chem, 2002. 277(22): p. 1931521. Xu, D., et al., Heparan sulfate regulates VEGF165 And VEGF121-mediated vascular hyperpermeability. J Biol Chem, 2010. Wijelath, E., et al., Multiple mechanisms for exogenous heparin modulation of vascular endothelial growth factor activity. J Cell Biochem, 2010. 111(2): p. 461-8. Zieris, A., et al., FGF-2 and VEGF functionalization of starPEG-heparin hydrogels to modulate biomolecular and physical cues of angiogenesis. Biomaterials, 2010. 31(31): p. 7985-94. Yang, H.S., et al., Delivery of basic fibroblast growth factor using heparinconjugated fibrin for therapeutic angiogenesis. Tissue Eng Part A, 2010. Woodruff, M.A., et al., Sustained release and osteogenic potential of heparan sulfate-doped fibrin glue scaffolds within a rat cranial model. J Mol Histol, 2007. 38(5): p. 425-33. Patil, S.D., F. Papadmitrakopoulos, and D.J. Burgess, Concurrent delivery of dexamethasone and VEGF for localized inflammation control and angiogenesis. J Control Release, 2007. 117(1): p. 68-79. Kivirikko, K.I. and J. Myllyharju, Prolyl 4-hydroxylases and their protein disulfide isomerase subunit. Matrix Biol, 1998. 16(7): p. 357-68. Hanauske-Abel, H.M., Prolyl 4-hydroxylase, a target enzyme for drug development. Design of suppressive agents and the in vitro effects of inhibitors and proinhibitors. J Hepatol, 1991. 13 Suppl 3: p. S8-15; discussion S16. Jaakkola, P., et al., Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science, 2001. 292(5516): p. 468-72. Lando, D., et al., FIH-1 is an asparaginyl hydroxylase enzyme that regulates the transcriptional activity of hypoxia-inducible factor. Genes Dev, 2002. 16(12): p. 1466-71. Hirsila, M., et al., Characterization of the human prolyl 4-hydroxylases that modify the hypoxia-inducible factor. J Biol Chem, 2003. 278(33): p. 30772-80. Kaelin, W.G., Proline hydroxylation and gene expression. Annu Rev Biochem, 2005. 74: p. 115-28. Bruick, R.K. and S.L. McKnight, Transcription. Oxygen sensing gets a second wind. Science, 2002. 295(5556): p. 807-8. Boudriot, U., et al., Electrospinning approaches toward scaffold engineering-a brief overview. Artif Organs, 2006. 30(10): p. 785-92. He, W., et al., Fabrication and endothelialization of collagen-blended biodegradable polymer nanofibers: potential vascular graft for blood vessel tissue engineering. Tissue Eng, 2005. 11(9-10): p. 1574-88. Li, D., J.T. McCann, and Y. Xia, Use of electrospinning to directly fabricate hollow nanofibers with functionalized inner and outer surfaces. Small, 2005. 1(1): p. 83-6. 152 212. 213. 214. 215. 216. 217. 218. 219. 220. 221. 222. 223. 224. 225. 226. 227. 228. 229. Cai, S., et al., Injectable glycosaminoglycan hydrogels for controlled release of human basic fibroblast growth factor. Biomaterials, 2005. 26(30): p. 605467. Prestwich, G.D., Simplifying the extracellular matrix for 3-D cell culture and tissue engineering: a pragmatic approach. J Cell Biochem, 2007. 101(6): p. 1370-83. Ekaputra, A.K., et al., Combining electrospun scaffolds with electrosprayed hydrogels leads to three-dimensional cellularization of hybrid constructs. Biomacromolecules, 2008. 9(8): p. 2097-103. Peattie, R.A., et al., Effect of gelatin on heparin regulation of cytokine release from hyaluronan-based hydrogels. Drug Deliv, 2008. 15(6): p. 389-97. Martinez, E.C., et al., Ascorbic Acid Improves Embryonic Cardiomyoblast Cell Survival and Promotes Vascularization in Potential Myocardial Grafts In Vivo. Tissue Eng Part A, 2010. Li, Y., et al., Direct labeling and visualization of blood vessels with lipophilic carbocyanine dye DiI. Nat Protoc, 2008. 3(11): p. 1703-8. Sung, H.J., et al., The effect of scaffold degradation rate on three-dimensional cell growth and angiogenesis. Biomaterials, 2004. 25(26): p. 5735-42. Keshaw, H., et al., Microporous collagen spheres produced via thermally induced phase separation for tissue regeneration. Acta Biomater, 2010. 6(3): p. 1158-66. Shen, X., et al., Prolyl hydroxylase inhibitors increase neoangiogenesis and callus formation following femur fracture in mice. J Orthop Res, 2009. 27(10): p. 1298-305. Ucuzian, A.A. and H.P. Greisler, In vitro models of angiogenesis. World J Surg, 2007. 31(4): p. 654-63. Leslie-Barbick, J.E., et al., Micron-scale Spatially Patterned, Covalently Immobilized VEGF on Hydrogels Accelerates Endothelial Tubulogenesis and Increases Cellular Angiogenic Responses. Tissue Eng Part A, 2010. Jia, X., et al., Sustained Release of VEGF by Coaxial Electrospun Dextran/PLGA Fibrous Membranes in Vascular Tissue Engineering. J Biomater Sci Polym Ed, 2010. Miyagi, Y., et al., Biodegradable collagen patch with covalently immobilized VEGF for myocardial repair. Biomaterials, 2010. Tschank, G., H.M. Hanauske-Abel, and B. Peterkofsky, The effectiveness of inhibitors of soluble prolyl hydroxylase against the enzyme in the cisternae of isolated bone microsomes. Arch Biochem Biophys, 1988. 261(2): p. 312-23. Pike, D.B., et al., Heparin-regulated release of growth factors in vitro and angiogenic response in vivo to implanted hyaluronan hydrogels containing VEGF and bFGF. Biomaterials, 2006. 27(30): p. 5242-51. Salzmann, D.L., et al., The effects of porosity on endothelialization of ePTFE implanted in subcutaneous and adipose tissue. J Biomed Mater Res, 1997. 34(4): p. 463-76. Pham, Q.P., U. Sharma, and A.G. Mikos, Electrospun poly(epsiloncaprolactone) microfiber and multilayer nanofiber/microfiber scaffolds: characterization of scaffolds and measurement of cellular infiltration. Biomacromolecules, 2006. 7(10): p. 2796-805. Baker, B.M., et al., The potential to improve cell infiltration in composite fiberaligned electrospun scaffolds by the selective removal of sacrificial fibers. Biomaterials, 2008. 29(15): p. 2348-58. 153 230. 231. 232. 233. 234. 235. 236. 237. Cao, H., et al., The topographical effect of electrospun nanofibrous scaffolds on the in vivo and in vitro foreign body reaction. J Biomed Mater Res A, 2010. 93(3): p. 1151-9. Leong, M.F., et al., Fabrication and in vitro and in vivo cell infiltration study of a bilayered cryogenic electrospun poly(D,L-lactide) scaffold. J Biomed Mater Res A, 2010. 94(4): p. 1141-9. Ju, Y.M., et al., Bilayered scaffold for engineering cellularized blood vessels. Biomaterials, 2010. 31(15): p. 4313-21. Anderson, J.M., Inflammatory response to implants. ASAIO Trans, 1988. 34(2): p. 101-7. Kutryk, M.J. and D.J. Stewart, Angiogenesis of the heart. Microsc Res Tech, 2003. 60(2): p. 138-58. Dvir, T., et al., Prevascularization of cardiac patch on the omentum improves its therapeutic outcome. Proc Natl Acad Sci U S A, 2009. 106(35): p. 149905. Leor, J., et al., Bioengineered cardiac grafts: A new approach to repair the infarcted myocardium? Circulation, 2000. 102(19 Suppl 3): p. III56-61. Madden, L.R., et al., Proangiogenic scaffolds as functional templates for cardiac tissue engineering. Proc Natl Acad Sci U S A, 2010. 107(34): p. 15211-6. 154 Appendix A.1 Standard curves for PHi drug release measurement Figure A Standard curves of PHi measured by spectophotometry. (a) PDCA was measured at an absorbance of 276 nm from a range of 0.01 mM to mM (0.37 μg to 27.75 μg) PDCA. (b) CPX was measured at an absorbance of 305 nm from a range of 0.1 μM to 25 μM (5.36 ng to 2144 ng) CPX. (c) HDZ was measured at an absorbance of 304 nm from a range of 1μM to 500 μM (40 ng to 20 μg) HDZ. a A.2 Publications 1. Lareu RR, Subramhanya KH, Peng Y, Benny P, Chen C, Wang Z, Rajagopalan R, Raghunath M. Collagen matrix deposition is dramatically enhanced in vitro when crowded with charged macromolecules: The biological relevance of the excluded volume effect. FEBS Lett. 2007 Jun 12;581(14):2709-2714. Epub 2007 May 21. 2. Chen ZC, Ekaputra AK, Gauthaman K, Adaikan PG, Yu H, Hutmacher DW. In vitro and in vivo analysis of co-electrospun scaffolds made of medical grade poly(epsilon-caprolactone) and porcine collagen. J Biomater Sci Polym Ed. 2008;19(5):693-707. 3. Wang Z, Chen C, Finger SN, Kwajah S, Jung M, Schwarz H, Swanson N, Lareu FF, Raghunath M: Suberoylanilide hydroxamic acid: a potential epigenetic therapeutic agent for lung fibrosis? Eur Respir J 2009, 34:145155. 4. Chen CZ, Peng YX, Wang ZB, Fish PV, Kaar JL, Koepsel RR, Russell AJ, Lareu RR, Raghunath M. The Scar-in-a-Jar: studying potential antifibrotic compounds from the epigenetic to extracellular level in a single well. British Journal of Pharmacology 2009, 158:1196-1209. 5. Chen CZC and Raghunath M. Focus on collagen: in vitro systems to study fibrogenesis and antifibrosis —state of the art. Fibrogenesis Tissue Repair 2009, 2(1):7 6. Chen C, Loe F, Blocki A, Peng Y and Raghunath M. Microenvironments by delegation: applying the principle of macromolecular crowding to enhance the extracellular matrix deposition and remodelling in vitro for tissue engineering and cell-based therapies. Advanced Drug Delivery Reviews 2010 (In press) b A.3 Successful grant applications written/participated in FRC—AcRF Tier Self vascularizing implant sensors—a pilot study. ($179,000) A.4 Awards 50 Best Abstracts Award. TERMIS-EU Meeting. June 13 – 16 Galway, Ireland, 2010 Clarice ZC Chen, Andrew K Ekaputra, Glenn D Prestwich, Michael Raghunath. Self-vascularizing Antifibrotic Biomaterials—A Pilot Study. (Oral presentation). c [...]... suggesting active import of proline into the wound [78] Providing additional proline or glutamine in the diet to enhance collagen biosynthesis, however, does not result in increased collagen accumulation In contrast, arginine, and ornithine supplementation are most effective in increasing collagen deposition [79] As cell culture media contains L-arginine and are usually supplemented with L-glutamine,... collagen molecule is composed of proline and hydroxyproline As a non-essential amino acid, proline is synthesized from arginine/ornithine via the urea cycle and glutamate (directly or indirectly from glutamine via glutaminase) through the citric acid cycle Clinical observations in burn patients suggest a drain of arginine, ornithine and glutamate [77], 19 while wound fluid proline levels are at least 50% higher... myocardium from dilatation and rupture, but on the other hand, it can impair cardiac function through increasing ventricular wall stiffness [17] Atherosclerotic lesions contain fibrotic tissue which can occupy up 87% of total plaque area [18] 12 Peri-implantational fibrosis represents a current clinical roadblock in regenerative medicine, which is gaining attention in the tissue engineering field Every implant... hydroxyproline in collagen Incorporation of the stable isotope of oxygen, 18O2, into collagen is also possible with this method and enables the examination of collagen synthesis in vitro [91] Polyacrylamide gel electrophoresis can be very collagen specific using metabolic labelling of cell cultures with radiolabelled amino acids glycine and proline [92] and subsequent detection of radioactive bands using... transforming growth factor 1 (TGF- 1) which supports wound healing and repair Under pathological conditions, TGF- 1 coordinates a cross-talk between parenchymal inflammatory and collagen-expressing cells, and plays a key role in fibrosis progression TGF- 1 is often referred to as a “soluble” factor We use quotation marks here because TGF- 1 is stored in its latent form bound to TGF- 1 binding proteins in. .. toxins, autoimmune and allergic reactions, radio- and chemotherapy can all lead to fibrosis This pathological process therefore can occur in almost any organ or tissue of the body, and typically results from situations persisting for several weeks or months in which inflammation, tissue destruction and repair occur simultaneously In this setting, fibrosis most frequently affects the lungs, liver, skin... the administration of hepatocyte growth factor [72] and matrix metalloproteinase 1 (MMP1) [57, 58] increases collagen turnover in the ECM It becomes clear that a meaningful in vitro system for the testing and characterisation of antifibrotics should be able to emulate the above described complete collagen matrix formation cascade, encompassing its biosynthesis and all post-translational (intra- and extra-cellular)... protein folding and protein-protein interactions In the case of fibrogenic cell culture, the conversion of procollagen to collagen is 21 sped up as well as the supramolecular assembly of collagen triple helices to form fibers 2.4.2 Quantitative issues—measuring collagen and normalising the data Determination of the amount of collagen produced in vitro is the next challenge, and this can be done in a... specificity in discriminating between collagen types, as well as a lack of internal normalization within samples, are disadvantages in a screening setting The Sirius dye has been used since 1964 to identify collagen in histology specimens [83, 84] It is based on the selective binding of Sirius Red F3BA to collagen Subsequent elution with sodium hydroxide-methanol and read-out at 540 nm can be done in cuvettes,... [44], and can in its active form be scavenged and possibly neutralized by decorin-mediated binding into the ECM [45, 46] (for review see [47]) Along with factors like epithelial growth factor, basic fibroblast growth factor and interleukin-1, TGF- 1 appears to play a key role in EMT [42] The connective tissue growth factor [48] and platelet-derived growth factor [49] have also been reported to be involved . Towards Topical Antifibrotics in Tissue Engineering and Repair Chen Zhen Cheng, Clarice (B. Appl. Sci Conclusion 71 Chapter 4 Peri-implantational fibrosis: a bottleneck in tissue engineering 72 4.1 Biomaterials in tissue engineering 73 4.2 Foreign body response 73 4.3 Biocompatibility 76 4.4. laboratories and R&D departments in the pharmaceutical industry have been facing in re-enacting the fibrotic process in vitro for screening procedures prior to animal testing. Effective in vitro

Ngày đăng: 10/09/2015, 15:52

TỪ KHÓA LIÊN QUAN