Applications of prolyl hydroxylase inhibitors in tissue engineering and regenerative medicine

142 261 0
Applications of prolyl hydroxylase inhibitors in tissue engineering and regenerative medicine

Đang tải... (xem toàn văn)

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

Thông tin tài liệu

APPLICATIONS OF PROLYL HYDROXYLASE INHIBITORS IN TISSUE ENGINEERING AND REGENERATIVE MEDICINE SHAM FONG WAI, ADELINE (B.Eng (Hons.), National University of Singapore) A THESIS SUBMITTED FOR THE DEGREE OF DOCTOR OF PHILOSOPHY DEPARTMENT OF BIOMEDICAL ENGINEERING NATIONAL UNIVERSITY OF SINGAPORE 2014 Acknowledgments I would like to express my deepest gratitude to my supervisor, Associate Professor Michael Raghunath, for his invaluable guidance and generous support throughout my PhD project He has taught me so many things over the years, from the intricacies of microscopy to critical thinking and presentation skills His great knowledge and insight have been most inspiring, and his sense of humor has often made dark times much more bearable Working with him has been a most wonderful and enriching experience, for which I am infinitely grateful I am also extremely grateful to Dr Sebastian Beyer, Dr Eliana C Martinez, Dr Clarice Chen, Dr Ping Yuan, Dr Dieter Trau and Professor Casey Chan for their generous guidance and assistance, as well as their endless patience I am also tremendously grateful to Miss Samantha de Witte for her invaluable contributions to the osteoblast branch of this work Special thanks go to my lovely friends and wonderful colleagues from the Tissue Modulation Laboratory, the Department of Biomedical Engineering and the NUS Tissue Engineering Programme for their constant support, advice, assistance and encouragement, without which I could not have survived this long journey Last but not least, I would also like to thank my parents for being the best parents a daughter could ever wish for i Publications and Conferences Publications: Sham A, Martinez EC, Beyer S, Trau DW, Raghunath M Incorporation of a prolyl hydroxylase inhibitor into scaffolds: a strategy for stimulating vascularization (Accepted into Tissue Engineering Part A) Sham A, De Witte SFH, Raghunath M Differential effects of prolyl hydroxylase inhibitors on early and late osteogenic differentiation (In preparation) Conferences: Sham A, Martinez EC, Beyer S, Trau DW, Raghunath M Stimulation of angiogenesis in tissue engineered constructs using prolyl hydroxylase inhibitors The 15th International Conference on Biomedical Engineering (ICBME), 4-7 December 2013, Singapore (Oral presentation) Sham A, Martinez EC, Beyer S, Trau DW, Raghunath M Incorporation of prolyl hydroxylase inhibitors into scaffolds: a strategy for stimulating vascularization Tissue Engineering and Regenerative Medicine International Society (TERMIS) Asia-Pacific 2013 Annual Conference, 23-26 October 2013, Shanghai/Wuzhen, China (Oral presentation) Awarded the First Prize for Best Oral Presentation ii Sham A, Beyer S, Trau DW, Raghunath M Engineering a proangiogenic and anti-fibrotic tissue engineering scaffold TERMIS World Congress 2012, 5-8 September 2012, Vienna, Austria (Poster presentation) Sham A, Beyer S, Martinez EC, Trau DW, Raghunath M Development of a pro-angiogenic and anti-fibrotic tissue engineering scaffold The International Union of Materials Research Societies - International Conference of Young Researchers on Advanced Materials 2012, 1-6 July 2012, Singapore (Poster presentation) Sham A, Chen C, Martinez EC, Ekaputra A, Beyer S, Prestwich GD, Trau DW, Raghunath M Pro-angiogenic and anti-fibrotic scaffolds for tissue engineering applications Keystone Symposia: Angiogenesis: Advances in Basic Science and Therapeutic Applications, 16-21 January 2012, Snowbird, Utah, USA (Poster presentation) Sham A, Chen C, Beyer S, Trau DW, Raghunath M Pharmacologic stimulation of angiogenesis and inhibition of fibrosis in tissueengineered constructs TERMIS Asia-Pacific 2011 Annual Conference, 3-5 August 2011, Singapore (Poster presentation) iii Table of Contents Acknowledgments i Publications and Conferences ii Table of Contents iv Summary vii List of Abbreviations ix List of Tables xi List of Figures xii Chapter Introduction 1.1 Background 1.1.1 Regenerative medicine – a new paradigm in healthcare 1.1.2 Vascularization is a major obstacle in tissue engineering 1.1.3 Current vascularization strategies for engineered tissues 1.2 Objectives and thesis scope 12 Chapter HIF-1 and PHIs in Angiogenesis 14 2.1 Overview of angiogenesis 15 2.2 HIF-1, PHIs and angiogenesis 18 2.2.1 HIF-1 structure and function 18 2.2.2 Molecular regulation of HIF-1 21 2.2.3 PHIs stimulate angiogenesis 24 2.3 Potential applications of PHIs 25 2.3.1 Ischemic and fibrotic diseases 25 2.3.2 Wound and fracture healing 28 2.4 Potential applications of PHIs in tissue engineering 31 Chapter Incorporation of a PHI into Scaffolds: A Vascularization Strategy for Tissue Engineering Applications 33 3.1 Introduction 34 3.2 Hypothesis and objectives 35 3.3 Materials and methods 35 3.3.1 Preparation of PDCA-Gelfoam 35 3.3.2 Drug loading measurements 38 3.3.3 Scanning electron microscopy 39 3.3.4 Cell culture 39 iv 3.3.5 Culturing fibroblasts on PDCA-Gelfoam scaffolds 40 3.3.6 Cytotoxicity assay 41 3.3.7 Quantifying cell numbers in scaffolds 41 3.3.8 Assessing the distribution of cells within the scaffolds 42 3.3.9 HIF-1α reporter assay 43 3.3.10 Analysis of VEGF secretion 43 3.3.11 Rat peri-renal fat implantation model 44 3.3.12 Preparation of frozen sections 45 3.3.13 Morphometric analysis of vascular infiltration 45 3.3.14 Statistical analysis 46 3.4 Results 46 3.4.1 PDCA was successfully conjugated to Gelfoam 46 3.4.2 Gelfoam retains high porosity after PDCA conjugation 47 3.4.3 PDCA-Gelfoam has low cytotoxicity and supports cell attachment, proliferation, and infiltration 50 3.4.4 HIF-1α is stabilized in a dose-dependent manner in cells growing on PDCA-Gelfoam 53 3.4.5 PDCA-Gelfoam stimulates VEGF secretion by fibroblasts in vitro 53 3.4.6 PDCA-Gelfoam stimulates vascular infiltration in vivo 56 3.5 Discussion 60 3.6 Conclusion 62 Chapter HIF-1 and the Potential Roles of PHIs in Bone Tissue Engineering and Regeneration 64 4.1 Introduction 65 4.2 Bone development and regeneration 65 4.2.1 Mechanisms of bone formation 65 4.2.2 Bone regeneration during fracture healing 66 4.3 The roles of HIF-1 in bone 69 4.3.1 HIF-1 and chondrocyte survival in hypoxia 69 4.3.2 HIF-1’s role in angiogenesis and osteogenesis 70 4.3.3 HIF-1 in osteogenic and chondrogenic differentiation 74 4.4 PHIs in bone regeneration 76 Chapter Effects of PHIs on Osteoblasts: A Preliminary Study 78 5.1 Introduction 79 5.2 Hypotheses and objectives 79 v 5.3 Materials and methods 80 5.3.1 Osteoblast culture 80 5.3.2 Preparation of PHIs for drug treatment 81 5.3.3 Preparation of fixatives 82 5.3.4 Cytotoxicity assay 82 5.3.5 Assessing PHIs’ effects on cellular HIF-1α levels 83 5.3.6 Durations of PHI treatment 83 5.3.7 Analysis of VEGF secretion 84 5.3.8 Assessing PHIs’ effects on collagen secretion 84 5.3.9 Immunocytochemical staining for type I collagen and osterix 85 5.3.10 Alizarin red staining 86 5.3.11 Statistical analysis 86 5.4 Results 87 5.4.1 PHIs stabilize HIF-1α in osteoblasts 87 5.4.2 PHIs stimulate VEGF secretion by osteoblasts 88 5.4.3 PHIs reduce collagen production by osteoblasts 90 5.4.4 PHIs increase osterix protein levels in osteoblasts 92 5.4.5 Effects of PHI-treatment on cell attachment 95 5.4.6 Cytotoxicity assay 98 5.4.7 PHIs’ effects on mineralization 100 5.5 Discussion 103 5.6 Conclusion 106 Chapter Conclusions and Future Work 108 6.1 Summary of key findings 109 6.2 Future work 110 6.2.1 Assessing functional vascularization 110 6.2.2 Applying our findings pertaining to PDCA-Gelfoam 112 6.2.3 Developing PHI-delivering materials for bone regeneration and tissue engineering 113 6.3 Conclusions 114 References 115 vi Summary Clinical applications of tissue engineering are constrained by the ability of the implanted construct to invoke vascularization in adequate extent and velocity To overcome the current limitations presented by local delivery of single angiogenic factors, we explored the incorporation of prolyl hydroxylase inhibitors (PHIs) into scaffolds as an alternative vascularization strategy PHIs are small molecule drugs which can stabilize the alpha subunit of hypoxia-inducible factor (HIF-1), a key transcription factor that regulates a variety of angiogenic mechanisms, via the inhibition of a family of HIF-regulating enzymes known as the HIF prolyl hydroxylases (HIF-PHDs) In this project, we conjugated the PHI pyridine-2,4-dicarboxylic acid (PDCA) via amide bonds to a gelatin sponge (Gelfoam®) Fibroblasts cultured on PDCA-Gelfoam were able to infiltrate and proliferate in these scaffolds while secreting significantly more vascular endothelial growth factor (VEGF) than cells grown on Gelfoam without PDCA Reporter cells expressing GFP-tagged HIF-1α exhibited dosedependent stabilization of this angiogenic transcription factor when growing within PDCA-Gelfoam constructs Subsequently, we implanted PDCA-Gelfoam scaffolds into the peri-renal fat tissue of Sprague Dawley rats for days Immunostaining of explants revealed that the PDCA-Gelfoam scaffolds were amply infiltrated by cells and promoted vascular ingrowth in a dose-dependent manner Thus, the vii incorporation of PHIs into scaffolds appears to be a feasible strategy for improving vascularization in regenerative medicine applications Aside from promoting angiogenesis, PHIs can also exert a range of other effects on cells and tissues As HIF-1 has been shown to be involved in bone development, PHIs’ applications in bone regeneration are of particular interest However, PHIs also inhibit collagen prolyl 4hydroxylase (P4H), and can thus suppress the production of collagen, an important component of bone Therefore, PHIs’ effects on bone are complex To explore PHIs’ effects on bone, we performed a preliminary study to investigate PHIs’ effects on several aspects of osteoblast behaviour in vitro, by treating osteoblasts with the PHIs PDCA, ciclopirox olamine (CPX), and desferrioxamine (DFO) Our results showed that all the tested PHIs could stabilize HIF-1α, upregulate VEGF secretion, and downregulate collagen secretion and deposition However, our results also revealed that different PHIs can have varied effects on osteoblast viability and mineralization, likely due to their different mechanisms of action and ranges of inhibitory targets We also showed that the duration of PHI treatment has an influence on resultant osteoblast behavior Taken together, our results suggest that a short initial treatment with non-iron chelator PHIs may be preferable in bone applications, although in vivo testing in suitable animal models of bone injury will be necessary before conclusions can be drawn regarding their efficacy viii List of Abbreviations ARNT: Aryl hydrocarbon receptor nuclear translocator bFGF: Basic fibroblast growth factor bHLH: Basic helix-loop-helix domain BSA: Bovine serum albumin CAD: Computer-aided design CBP: CREB-binding protein CDI: 1,1’-Carbonyldiimidazole CPX: Ciclopirox olamine DAPI: 4',6-Diamidino-2-phenylindole DFO: Desferrioxamine DMEM: Dulbecco's Modified Eagle Medium DMOG: Dimethyloxalylglycine EC: Endothelial cell ELISA: Enzyme-linked immunosorbent assay FBS: Fetal bovine serum FIH: Factor inhibiting HIF-1 Flt-1: Fms-like tyrosine kinase G6PD: Glucose 6-phosphate dehydrogenase GLUT1: Glucose transporter GMP: Good manufacturing practices HDZ: Hydralazine hydrochloride HGF: Hepatocyte growth factor HIF-1: Hypoxia-inducible factor ix volume, average vessel diameter, and degree of anisotropy [154] Vessels in the three-dimensional reconstruction can also be colorcoded by vessel diameter [154] 6.2.2 Applying our findings pertaining to PDCA-Gelfoam Soft tissue engineering applications As described in the previous section, we have developed a method to incorporate a PHI into amine-containing scaffolds and demonstrated its feasibility in stimulating vascularization Our proof-of-concept scaffold, PDCA-Gelfoam, was conducive to cell attachment, proliferation, and infiltration, and was able to induce vascular infiltration in a dosedependent manner However, as the base material (Gelfoam) is a gelatin sponge, the PDCA-Gelfoam scaffolds have relatively low mechanical strength and may be suitable mainly for soft tissue engineering applications Studies can thus be performed to assess PDCA-Gelfoam’s efficacy in accelerating tissue regeneration in animal models for specific applications (e.g chronic wounds, myocardial infarction) As our method of incorporating PDCA into scaffolds utilizes PDCA’s intrinsic carboxylic acid groups to form amide bonds with amine groups in the scaffold, it is compatible with all amine-containing materials These include all protein-based materials, as well as synthetic polymers containing amine groups, such as nylon and other 112 polyamides Therefore, depending on the needs of the specific application, the material can be switched from Gelfoam to any of these other materials to match the needs Combining PDCA-Gelfoam with in vitro pre-vascularization In our in vivo study, we explored PDCA-Gelfoam’s ability to stimulate vascularization as a standalone material (i.e without pre-seeding it with cells before implantation), and showed that it was effective on its own However, since the incorporated PDCA stimulates vascularization by switching on the intrinsic angiogenic programming in infiltrating cells, it should also be able to improve in vitro pre-vascularization by similarly enhancing the formation of capillary pre-cursors The incorporation of PHIs into scaffolds is thus likely to work synergistically with in vitro prevascularization to further improve vascular infiltration, and we should try combining the two techniques in future studies 6.2.3 Developing PHI-delivering materials for bone regeneration and tissue engineering As reviewed in chapter 3, PHIs have many potential applications in bone and cartilage regeneration Although PDCA-Gelfoam exhibited excellent compatibility with cells and successfully stimulated vascularization in vivo, its lack of mechanical strength makes it unsuitable for applications in bone, since a scaffold should ideally have mechanical properties that match the tissue in which it is implanted, 113 and bone has important load-bearing functions [155] Therefore, different PHI-delivering scaffolds should be developed for bone tissue engineering applications Our preliminary studies in osteoblasts have also shown that different PHIs and treatment durations can have very different effects on the same cell type Therefore, it may be preferable to first test out multiple PHIs in appropriate animal models to determine the best PHI treatment, before designing a PHI-delivering material for a specific application 6.3 Conclusions In this PhD project, we have developed a simple and cost-effective method to incorporate PHIs into amine-containing scaffolds, and demonstrated that it is feasible as a strategy for improving vascular infiltration We have also explored the effects of three PHIs on osteoblasts, and found that while all the PHIs we tested can switch on HIF-1 signaling and decrease collagen production, the type of PHI used and the duration of treatment are also important factors influencing subsequent osteoblast behavior In conclusion, our results suggest that the use of PHIs is a promising approach for inducing vascularization in regenerating tissues Additionally, as different PHIs can have a wide range of effects in different tissues, more studies will have to be performed in suitable animal models to determine their efficacy in specific applications 114 References Kirkwood T.B (2008) A systematic look at an old problem Nature 451, 644-647 Centers for Disease Control and Prevention (1999) Achievements in Public Health, 1900-1999: Control of Infectious Diseases Retrieved on January 6, 2014 from the website: http://www.cdc.gov/mmwr/preview/mmwrhtml/mm4829a1.htm World Health Organization (2014) Chronic diseases and health promotion Retrieved on January 2014 from the website: http://www.who.int/chp/en/ Michaels P.J., Fishbein M.C., Colvin R.B (2003) Humoral rejection of human organ transplants Springer Semin Immunopathol 25, 119140 Feng S., Buell J.F., Cherikh W.S., et al (2002) Organ donors with positive viral serology or malignancy: risk of disease transmission by transplantation Transplantation 74, 1657-1663 Abouna G.M (2008) Organ shortage crisis: problems and possible solutions Transplant Proc 40, 34-38 United States Department of Health and Human Services (2014) OPTN: Organ Procurement and Transplantation Network Retrieved on January 6, 2014 from the website: http://optn.transplant.hrsa.gov/ Chapekar M.S (2000) Tissue engineering: challenges and opportunities J Biomed Mater Res 53, 617-620 Huet, N (2013) Reuters: France's Carmat implants its first artificial heart in human Retrieved on January 6, 2014 from the website: http://www.reuters.com/article/2013/12/20/us-carmat-implantidUSBRE9BJ11L20131220 10 Langer R., Vacanti J.P (1993) Tissue engineering Science 260, 920-926 11 Daar A.S., Greenwood H.L (2007) A proposed definition of regenerative medicine J Tissue Eng Regen Med 1, 179-184 12 Mason C., Dunnill P (2008) A brief definition of regenerative medicine Regen Med 3, 1-5 13 Johnson P.C., Mikos A.G., Fisher J.P., et al (2007) Strategic directions in tissue engineering Tissue Eng 13, 2827-2837 115 14 Novosel E.C., Kleinhans C., Kluger P.J (2011) Vascularization is the key challenge in tissue engineering Adv Drug Deliv Rev 63, 300311 15 Naderi H., Matin M.M., Bahrami A.R (2011) Review paper: critical issues in tissue engineering: biomaterials, cell sources, angiogenesis, and drug delivery systems J Biomater Appl 26, 383-417 16 Rouwkema J., Rivron N.C., van Blitterswijk C.A (2008) Vascularization in tissue engineering Trends Biotechnol 26, 434-441 17 Carmeliet P., Jain R.K (2000) Angiogenesis in cancer and other diseases Nature 407, 249-257 18 Koffler J., Kaufman-Francis K., Shandalov Y., et al (2011) Improved vascular organization enhances functional integration of engineered skeletal muscle grafts Proc Natl Acad Sci U S A 108, 14789-14794 19 Ring A., Langer S., Tilkorn D., et al (2010) Induction of angiogenesis and neovascularization in adjacent tissue of plasmacollagen-coated silicone implants Eplasty 10, e61 20 Adams R.H., Alitalo K (2007) Molecular regulation of angiogenesis and lymphangiogenesis Nat Rev Mol Cell Biol 8, 464-478 21 Plopper G (2012) Principles of Cell Biology Burlington, MA: Jones & Bartlett Learning, pp 205-6 22 Pierschbacher M.D., Ruoslahti E (1984) Cell attachment activity of fibronectin can be duplicated by small synthetic fragments of the molecule Nature 309, 30-33 23 Hersel U., Dahmen C., Kessler H (2003) RGD modified polymers: biomaterials for stimulated cell adhesion and beyond Biomaterials 24, 4385-4415 24 Druecke D., Langer S., Lamme E., et al (2004) Neovascularization of poly(ether ester) block-copolymer scaffolds in vivo: long-term investigations using intravital fluorescent microscopy J Biomed Mater Res A 68, 10-18 25 Karageorgiou V., Kaplan D (2005) Porosity of 3D biomaterial scaffolds and osteogenesis Biomaterials 26, 5474-5491 26 Yang S., Leong K.F., Du Z., et al (2001) The design of scaffolds for use in tissue engineering Part I Traditional factors Tissue Eng 7, 679-689 27 Hutmacher D.W., Sittinger M., Risbud M.V (2004) Scaffold-based tissue engineering: rationale for computer-aided design and solid freeform fabrication systems Trends Biotechnol 22, 354-362 116 28 Hollister S.J (2005) Porous scaffold design for tissue engineering Nat Mater 4, 518-524 29 Lantada A.D., Morgado P.L (2012) Rapid prototyping for biomedical engineering: current capabilities and challenges Annu Rev Biomed Eng 14, 73-96 30 Nomi M., Atala A., Coppi P.D., et al (2002) Principals of neovascularization for tissue engineering Mol Aspects Med 23, 463483 31 Hubbell J.A (1999) Bioactive biomaterials Curr Opin Biotech 10, 123-129 32 Papanas N., Maltezos E (2010) Benefit-risk assessment of becaplermin in the treatment of diabetic foot ulcers Drug Saf 33, 455461 33 Kneser U., Polykandriotis E., Ohnolz J., et al (2006) Engineering of vascularized transplantable bone tissues: induction of axial vascularization in an osteoconductive matrix using an arteriovenous loop Tissue Eng 12, 1721-1731 34 Levenberg S., Rouwkema J., Macdonald M., et al (2005) Engineering vascularized skeletal muscle tissue Nat Biotechnol 23, 879-884 35 Tremblay P.L., Hudon V., Berthod F., et al (2005) Inosculation of Tissue ‐ Engineered Capillaries with the Host's Vasculature in a Reconstructed Skin Transplanted on Mice Am J Transplant 5, 10021010 36 Lovett M., Lee K., Edwards A., et al (2009) Vascularization strategies for tissue engineering Tissue Eng Part B Rev 15, 353-370 37 Zisch A.H., Lutolf M.P., Hubbell J.A (2003) Biopolymeric delivery matrices for angiogenic growth factors Cardiovasc Pathol 12, 295310 38 Zhao W., Han Q., Lin H., et al (2008) Improved neovascularization and wound repair by targeting human basic fibroblast growth factor (bFGF) to fibrin J Mol Med (Berl) 86, 1127-1138 39 Andrae J., Gallini R., Betsholtz C (2008) Role of platelet-derived growth factors in physiology and medicine Genes Dev 22, 1276-1312 40 Shea L.D., Smiley E., Bonadio J., et al (1999) DNA delivery from polymer matrices for tissue engineering Nat Biotechnol 17, 551-554 41 Unger R.E., Halstenberg S., Sartoris A., et al (2011) Human endothelial and osteoblast co-cultures on 3D biomaterials Methods Mol Biol 695, 229-241 117 42 Epstein A.C., Gleadle J.M., McNeill L.A., et al (2001) C elegans EGL-9 and Mammalian Homologs Define a Family of Dioxygenases that Regulate HIF by Prolyl Hydroxylation Cell 107, 43-54 43 Tschank G., Raghunath M., Gunzler V., et al (1987) 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 248, 625-633 44 Viguet-Carrin S., Garnero P., Delmas P.D (2006) The role of collagen in bone strength Osteoporos Int 17, 319-336 45 Adair T.H., Montani J.P (2011) Angiogenesis San Rafael, CA: Morgan & Claypool Life Sciences, pp 1-8 46 Flamme I., Frolich T., Risau W (1997) Molecular mechanisms of vasculogenesis and embryonic angiogenesis J Cell Physiol 173, 206210 47 Leslie J.D., Ariza-McNaughton L., Bermange A.L., et al (2007) Endothelial signalling by the Notch ligand Delta-like restricts angiogenesis Development 134, 839-844 48 Siekmann A.F., Lawson N.D (2007) Notch signalling limits angiogenic cell behaviour in developing zebrafish arteries Nature 445, 781-784 49 Geudens I., Gerhardt H (2011) Coordinating cell behaviour during blood vessel formation Development 138, 4569-4583 50 Ke Q., Costa M (2006) Hypoxia-inducible factor-1 (HIF-1) Mol Pharmacol 70, 1469-1480 51 Wang G.L., Jiang B.H., Rue E.A., et al (1995) Hypoxia-inducible factor is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension Proc Natl Acad Sci U S A 92, 5510-5514 52 Lando D., Peet D.J., Gorman J.J., et al (2002) FIH-1 is an asparaginyl hydroxylase enzyme that regulates the transcriptional activity of hypoxia-inducible factor Genes Dev 16, 1466-1471 53 Maes C., Carmeliet G., Schipani E (2012) Hypoxia-driven pathways in bone development, regeneration and disease Nat Rev Rheumatol 8, 358-366 54 Semenza G.L (2003) Targeting HIF-1 for cancer therapy Nat Rev Cancer 3, 721-732 55 Schofield C.J., Ratcliffe P.J (2004) Oxygen sensing by HIF hydroxylases Nat Rev Mol Cell Biol 5, 343-354 118 56 Minchenko A., Salceda S., Bauer T., et al (1993) Hypoxia regulatory elements of the human vascular endothelial growth factor gene Cell Mol Biol Res 40, 35-39 57 Liu Y., Cox S.R., Morita T., et al (1995) Hypoxia regulates vascular endothelial growth factor gene expression in endothelial cells identification of a 5′ enhancer Circ Res 77, 638-643 58 Levy A.P., Levy N.S., Wegner S., et al (1995) Transcriptional regulation of the rat vascular endothelial growth factor gene by hypoxia J Biol Chem 270, 13333-13340 59 LeCouter J., Kowalski J., Foster J., et al (2001) Identification of an angiogenic mitogen selective for endocrine gland endothelium Nature 412, 877-884 60 Scheid A., Wenger R.H., Schäffer L., et al (2002) Physiologically low oxygen concentrations in fetal skin regulate hypoxia-inducible factor and transforming growth factor-β3 FASEB J 16, 411-413 61 Gerber H.-P., Condorelli F., Park J., et al (1997) Differential transcriptional regulation of the two vascular endothelial growth factor receptor genes Flt-1, but not Flk-1/KDR, is up-regulated by hypoxia J Biol Chem 272, 23659-23667 62 Hanauske-Abel H., Günzler V (1982) A stereochemical concept for the catalytic mechanism of prolylhydroxylase: applicability to classification and design of inhibitors J Theor Biol 94, 421-455 63 Hirsila M., Koivunen P., Gunzler V., et al (2003) Characterization of the human prolyl 4-hydroxylases that modify the hypoxia-inducible factor J Biol Chem 278, 30772-30780 64 Ivan M., Haberberger T., Gervasi D.C., et al (2002) Biochemical purification and pharmacological inhibition of a mammalian prolyl hydroxylase acting on hypoxia-inducible factor Proc Natl Acad Sci U S A 99, 13459-13464 65 Shoulders M.D., Raines R.T (2009) Collagen structure and stability Annu Rev Biochem 78, 929-958 66 Fraisl P., Aragones J., Carmeliet P (2009) Inhibition of oxygen sensors as a therapeutic strategy for ischaemic and inflammatory disease Nat Rev Drug Discov 8, 139-152 67 Muchnik E., Kaplan J (2011) HIF prolyl hydroxylase inhibitors for anemia Expert Opin Investig Drugs 20, 645-656 68 Warnecke C., Griethe W., Weidemann A., et al (2003) Activation of the hypoxia-inducible factor-pathway and stimulation of angiogenesis by application of prolyl hydroxylase inhibitors FASEB J 17, 11861188 119 69 Raghunath M., Wong Y.S., Farooq M., et al (2009) Pharmacologically induced angiogenesis in transgenic zebrafish Biochem Biophys Res Commun 378, 766-771 70 Semenza G.L (2001) Hypoxia-inducible factor 1: oxygen homeostasis and disease pathophysiology Trends Mol Med 7, 345350 71 Franklin T.J (1997) Therapeutic approaches to organ fibrosis Int J Biochem Cell Biol 29, 79-89 72 Ockaili R., Natarajan R., Salloum F., et al (2005) HIF-1 activation attenuates postischemic myocardial injury: role for heme oxygenase-1 in modulating microvascular chemokine generation Am J Physiol Heart Circ Physiol 289, H542-548 73 Natarajan R., Salloum F.N., Fisher B.J., et al (2006) Hypoxia inducible factor-1 activation by prolyl 4-hydroxylase-2 gene silencing attenuates myocardial ischemia reperfusion injury Circ Res 98, 133140 74 Nwogu J.I., Geenen D., Bean M., et al (2001) Inhibition of Collagen Synthesis With Prolyl 4-Hydroxylase Inhibitor Improves Left Ventricular Function and Alters the Pattern of Left Ventricular Dilatation After Myocardial Infarction Circulation 104, 2216-2221 75 Kim I., Mogford J.E., Witschi C., et al (2003) Inhibition of prolyl 4hydroxylase reduces scar hypertrophy in a rabbit model of cutaneous scarring Wound Repair Regen 11, 368-372 76 Fujiwara K., Ogata I., Ohta Y., et al (1988) Decreased collagen accumulation by a prolyl hydroxylase inhibitor in pig serum-induced fibrotic rat liver Hepatology 8, 804-807 77 Bickel M., Baringhaus K.H., Gerl M., et al (1998) Selective inhibition of hepatic collagen accumulation in experimental liver fibrosis in rats by a new prolyl 4-hydroxylase inhibitor Hepatology 28, 404411 78 Siddiq A., Ayoub I.A., Chavez J.C., et al (2005) Hypoxia-inducible factor prolyl 4-hydroxylase inhibition A target for neuroprotection in the central nervous system J Biol Chem 280, 41732-41743 79 Haase V.H (2006) The VHL/HIF oxygen-sensing pathway and its relevance to kidney disease Kidney Int 69, 1302-1307 80 Bernhardt W.M., Campean V., Kany S., et al (2006) Preconditional activation of hypoxia-inducible factors ameliorates ischemic acute renal failure J Am Soc Nephrol 17, 1970-1978 120 81 Finkelstein E.A., Corso P.S., Miller T.R (2006) Incidence and Economic Burden of Injuries in the United States New York, NY: Oxford University Press, pp 69 82 Malay D.S., Margolis D.J., Hoffstad O.J., et al (2006) The incidence and risks of failure to heal after lower extremity amputation for the treatment of diabetic neuropathic foot ulcer J Foot Ankle Surg 45, 366-374 83 Steinmann S.P., Adams J.E (2006) Scaphoid fractures and nonunions: diagnosis and treatment J Orthop Sci 11, 424-431 84 Shen X., Wan C., Ramaswamy G., et al (2009) Prolyl hydroxylase inhibitors increase neoangiogenesis and callus formation following femur fracture in mice J Orthop Res 27, 1298-1305 85 Schipani E., Maes C., Carmeliet G., et al (2009) Regulation of osteogenesis-angiogenesis coupling by HIFs and VEGF J Bone Miner Res 24, 1347-1353 86 Wan C., Gilbert S.R., Wang Y., et al (2008) Activation of the hypoxia-inducible factor-1alpha pathway accelerates bone regeneration Proc Natl Acad Sci U S A 105, 686-691 87 Rabinowitz M.H., Barrett T.D., Rosen M.D., et al (2010) Inhibitors of HIF Prolyl Hydroxylases Annu Rep Med Chem 45, 123 88 Thangarajah H., Vial I.N., Grogan R.H., et al (2010) HIF-1alpha dysfunction in diabetes Cell Cycle 9, 75-79 89 Thangarajah H., Yao D., Chang E.I., et al (2009) The molecular basis for impaired hypoxia-induced VEGF expression in diabetic tissues Proc Natl Acad Sci U S A 106, 13505-13510 90 Ko S.H., Nauta A., Morrison S.D., et al (2011) Antimycotic ciclopirox olamine in the diabetic environment promotes angiogenesis and enhances wound healing PLoS One 6, e27844 91 Saiki R.K., Gelfand D.H., Stoffel S., et al (1988) Primer-directed enzymatic amplification of DNA with a thermostable DNA polymerase Science 239, 487-491 92 Martinez E.C., Wang J., Gan S.U., et al (2010) Ascorbic acid improves embryonic cardiomyoblast cell survival and promotes vascularization in potential myocardial grafts in vivo Tissue Eng Part A 16, 1349-1361 93 Stephenson H.P., Sponer H (1957) Near Ultraviolet Absorption Spectra of the Pyridine Monocarboxylic Acids in Water and Ethanol Solutions1, J Am Chem Soc 79, 2050-2056 121 94 Banga A.K (2005) Therapeutic Peptides and Proteins: Formulation, Processing, and Delivery Systems, Second Edition Boca Raton, FL: Taylor & Francis, pp 43 95 Chen C., Peng Y., Wang Z., et al (2009) The Scar‐in‐a‐Jar: studying potential antifibrotic compounds from the epigenetic to extracellular level in a single well Brit J Pharmacol 158, 1196-1209 96 Agis H., Watzek G., Gruber R (2012) Prolyl hydroxylase inhibitors increase the production of vascular endothelial growth factor by periodontal fibroblasts J Periodontal Res 47, 165-173 97 Wong Y.S (2008) Applying side effects of anti fibrotic compounds to promote neovascularization for tissue engineering [PhD Thesis] Division of Bioengineering, National University of Singapore, Singapore, 2008 98 Finn M.D., Schow S.R., Schneiderman E.D (1992) Osseous regeneration in the presence of four common hemostatic agents J Oral Maxillofac Surg 50, 608-612 99 Dahm M., Lyman W.D., Schwell A.B., et al (1990) Immunogenicity of glutaraldehyde-tanned bovine pericardium J Thorac Cardiovasc Surg 99, 1082-1090 100 Madhavan K., Belchenko D., Motta A., et al (2010) Evaluation of composition and crosslinking effects on collagen-based composite constructs Acta Biomater 6, 1413-1422 101 Wan C., Shao J., Gilbert S.R., et al (2010) Role of HIF-1alpha in skeletal development Ann N Y Acad Sci 1192, 322-326 102 Schipani E (2005) Hypoxia and HIF-1 alpha in chondrogenesis Semin Cell Dev Biol 16, 539-546 103 Ong S.G., Hausenloy D.J (2012) Hypoxia-inducible factor as a therapeutic target for cardioprotection Pharmacol Ther 136, 69-81 104 Cai Z., Manalo D.J., Wei G., et al (2003) Hearts from rodents exposed to intermittent hypoxia or erythropoietin are protected against ischemia-reperfusion injury Circulation 108, 79-85 105 Natarajan R., Salloum F.N., Fisher B.J., et al (2008) Hypoxia inducible factor-1 upregulates adiponectin in diabetic mouse hearts and attenuates post-ischemic injury J Cardiovasc Pharm 51, 178-187 106 Schipani E., Khatri R (2010) The Role of Hypoxia-Induced Factors In: Bronner F., Farach-Carson M.C., Roach H.I., Bone and Development London: Springer-Verlag, pp 107-123 122 107 Safadi F.F., Barbe M.F., Abdelmagid S.M., et al (2009) Bone Structure, Development and Bone Biology In: Khurana J.S., Bone Pathology 2nd ed New York, NY: Springer, pp 16-17 108 Claes L., Recknagel S., Ignatius A (2012) Fracture healing under healthy and inflammatory conditions Nat Rev Rheumatol 8, 133-143 109 Claes L.E., Heigele C.A (1999) Magnitudes of local stress and strain along bony surfaces predict the course and type of fracture healing J Biomech 32, 255-266 110 Claes L (2011) Biomechanical principles and mechanobiologic aspects of flexible and locked plating J Orthop Trauma 25 Suppl 1, S4-7 111 Wallace A.L., Draper E.R., Strachan R.K., et al (1994) The vascular response to fracture micromovement Clin Orthop Relat Res 281-290 112 Claes L., Eckert-Hubner K., Augat P (2002) The effect of mechanical stability on local vascularization and tissue differentiation in callus healing J Orthop Res 20, 1099-1105 113 Kolar P., Schmidt-Bleek K., Schell H., et al (2010) The early fracture hematoma and its potential role in fracture healing Tissue Eng Part B Rev 16, 427-434 114 Aho A.J (1966) Electron microscopic and histological observations on fracture repair in young and old rats Acta Pathol Microbiol Scand Suppl 184:181-195 115 Megas P (2005) Classification of non-union Injury 36 Suppl 4, S30-37 116 Rüedi T.P., Murphy W.M (2000) AO Principles of Fracture Management Thieme, Stuttgart: AO Publishing, pp 731 117 Hayda R.A., Brighton C.T., Esterhai J.L., Jr (1998) Pathophysiology of delayed healing Clin Orthop Relat Res S31-40 118 Araldi E., Schipani E (2010) Hypoxia, HIFs and bone development Bone 47, 190-196 119 Zelzer E., Mamluk R., Ferrara N., et al (2004) VEGFA is necessary for chondrocyte survival during bone development Development 131, 2161-2171 120 Iyer N.V., Kotch L.E., Agani F., et al (1998) Cellular and developmental control of O2 homeostasis by hypoxia-inducible factor alpha Genes Dev 12, 149-162 123 121 Seagroves T.N., Ryan H.E., Lu H., et al (2001) Transcription factor HIF-1 is a necessary mediator of the pasteur effect in mammalian cells Mol Cell Biol 21, 3436-3444 122 Seagroves T., Johnson R.S (2002) Two HIFs may be better than one Cancer Cell 1, 211-213 123 Semenza G.L., Roth P.H., Fang H.M., et al (1994) Transcriptional regulation of genes encoding glycolytic enzymes by hypoxia-inducible factor J Biol Chem 269, 23757-23763 124 Kim J.W., Tchernyshyov I., Semenza G.L., et al (2006) HIF-1mediated expression of pyruvate dehydrogenase kinase: a metabolic switch required for cellular adaptation to hypoxia Cell Metab 3, 177185 125 Fukuda R., Zhang H., Kim J.W., et al (2007) HIF-1 regulates cytochrome oxidase subunits to optimize efficiency of respiration in hypoxic cells Cell 129, 111-122 126 Riddle R.C., Khatri R., Schipani E., et al (2009) Role of hypoxiainducible factor-1alpha in angiogenic-osteogenic coupling J Mol Med (Berl) 87, 583-590 127 Wang Y., Wan C., Deng L., et al (2007) The hypoxia-inducible factor alpha pathway couples angiogenesis to osteogenesis during skeletal development J Clin Invest 117, 1616-1626 128 Mayr-Wohlfart U., Waltenberger J., Hausser H., et al (2002) Vascular endothelial growth factor stimulates chemotactic migration of primary human osteoblasts Bone 30, 472-477 129 Deckers M.M., van Bezooijen R.L., van der Horst G., et al (2002) Bone morphogenetic proteins stimulate angiogenesis through osteoblast-derived vascular endothelial growth factor A Endocrinology 143, 1545-1553 130 Fiedler J., Leucht F., Waltenberger J., et al (2005) VEGF-A and PlGF-1 stimulate chemotactic migration of human mesenchymal progenitor cells Biochem Biophys Res Commun 334, 561-568 131 Street J., Lenehan B (2009) Vascular endothelial growth factor regulates osteoblast survival - evidence for an autocrine feedback mechanism J Orthop Surg Res 4, 19 132 Mayer H., Bertram H., Lindenmaier W., et al (2005) Vascular endothelial growth factor (VEGF-A) expression in human mesenchymal stem cells: autocrine and paracrine role on osteoblastic and endothelial differentiation J Cell Biochem 95, 827-839 133 Amarilio R., Viukov S.V., Sharir A., et al (2007) HIF1alpha regulation of Sox9 is necessary to maintain differentiation of hypoxic 124 prechondrogenic cells during early skeletogenesis Development 134, 3917-3928 134 Nakashima K., Zhou X., Kunkel G., et al (2002) The novel zinc finger-containing transcription factor osterix is required for osteoblast differentiation and bone formation Cell 108, 17-29 135 Zhou X., Zhang Z., Feng J.Q., et al (2010) Multiple functions of Osterix are required for bone growth and homeostasis in postnatal mice Proc Natl Acad Sci U S A 107, 12919-12924 136 Zhang C., Tang W., Li Y., et al (2011) Osteoblast-specific transcription factor Osterix increases vitamin D receptor gene expression in osteoblasts PLoS One 6, e26504 137 Bi W., Deng J.M., Zhang Z., et al (1999) Sox9 is required for cartilage formation Nat Genet 22, 85-89 138 Wagner T., Wirth J., Meyer J., et al (1994) Autosomal sex reversal and campomelic dysplasia are caused by mutations in and around the SRY-related gene SOX9 Cell 79, 1111-1120 139 Akiyama H., Lyons J.P., Mori-Akiyama Y., et al (2004) Interactions between Sox9 and beta-catenin control chondrocyte differentiation Genes Dev 18, 1072-1087 140 Feinberg R.N., Latker C.H., Beebe D.C (1986) Localized vascular regression during limb morphogenesis in the chicken embryo I Spatial and temporal changes in the vascular pattern Anat Rec 214, 405-409 141 Hallmann R., Feinberg R.N., Latker C.H., et al (1987) Regression of blood vessels precedes cartilage differentiation during chick limb development Differentiation 34, 98-105 142 Kolk A., Handschel J., Drescher W., et al (2012) Current trends and future perspectives of bone substitute materials - from space holders to innovative biomaterials J Craniomaxillofac Surg 40, 706718 143 Laurencin C.T (2003) Bone Graft Substitutes Conshohocken, PA: ASTM International, pp 11 West 144 Santos M.I., Reis R.L (2010) Vascularization in bone tissue engineering: physiology, current strategies, major hurdles and future challenges Macromol Biosci 10, 12-27 145 Gerstenfeld L.C., Chipman S.D., Kelly C.M., et al (1988) Collagen expression, ultrastructural assembly, and mineralization in cultures of chicken embryo osteoblasts J Cell Biol 106, 979-989 125 146 Shim S.H., Hah J.H., Hwang S.Y., et al (2010) Dexamethasone treatment inhibits VEGF production via suppression of STAT3 in a head and neck cancer cell line Oncol Rep 23, 1139-1143 147 Hegeman M.A., Hennus M.P., Cobelens P.M., et al (2013) Dexamethasone attenuates VEGF expression and inflammation but not barrier dysfunction in a murine model of ventilator-induced lung injury PLoS One 8, e57374 148 Cammack R., Wrigglesworth J.M., Baum H (1990) Irondependent Enzymes in Mammalian Systems In: Ponka P., Schulman H.M., Woodworth R.C., Richter G.W., Iron Transport and Storage Boca Raton, FL: Taylor & Francis, pp 17-39 149 Tschank G., Brocks D., Engelbart K., et al (1991) Inhibition of prolyl hydroxylation and procollagen processing in chick-embryo calvaria by a derivative of pyridine-2, 4-dicarboxylate Characterization of the diethyl ester as a proinhibitor Biochem J 275, 469-476 150 Eberhard Y., McDermott S.P., Wang X., et al (2009) Chelation of intracellular iron with the antifungal agent ciclopirox olamine induces cell death in leukemia and myeloma cells Blood 114, 3064-3073 151 Li Y., Song Y., Zhao L., et al (2008) Direct labeling and visualization of blood vessels with lipophilic carbocyanine dye DiI Nat Protoc 3, 1703-1708 152 Ceradini D.J., Kulkarni A.R., Callaghan M.J., et al (2004) Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1 Nat Med 10, 858-864 153 Jorgensen S.M., Demirkaya O., Ritman E.L (1998) Threedimensional imaging of vasculature and parenchyma in intact rodent organs with X-ray micro-CT Am J Physiol 275, H1103-1114 154 Duvall C.L., Taylor W.R., Weiss D., et al (2004) Quantitative microcomputed tomography analysis of collateral vessel development after ischemic injury Am J Physiol Heart Circ Physiol 287, H302-310 155 Hutmacher D.W (2000) Scaffolds in tissue engineering bone and cartilage Biomaterials 21, 2529-2543 126 ... for Tissue Engineering Applications 33 3.1 Introduction As explained in chapter 1, inadequate vascularization is a major bottleneck limiting widespread clinical applications of tissue engineering, ... artificial tissue and organ replacements This is the field of tissue engineering, which has been defined as "an interdisciplinary field that applies the principles of engineering and the life... regenerating human cells, tissues, and organs and restoring their function, while the term ? ?tissue engineering? ?? refers to the subset of regenerative medicine approaches that involve the design of tissue

Ngày đăng: 09/09/2015, 11:11

Từ khóa liên quan

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan