Tissue engineering of a vascularized bone graft

146 425 0
Tissue engineering of a vascularized bone graft

Đang tải... (xem toàn văn)

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

Thông tin tài liệu

TISSUE E GI EERI G OF A VASCULARIZED BO E GRAFT SUBHA ARAYA RATH (MBBS, MMST) A THESIS SUBMITTED FOR THE DEGREE OF DOCTOR OF PHILOSOPHY DIVISIO OF BIOE GI EERI G ATIO AL U IVERSITY OF SI GAPORE 2009 Acknowledgements It is a pleasure to thank those who made this thesis possible and the list cannot be completed with few names. I understood at the end of my PhD work that the journey of few years was possible only with the help of the innumerable number of friends, teachers and mentors whose unique help was absolutely important in maintaining proper direction. Therefore, first of all, I would like to thank all those persons who came across in my PhD journey and their contribution was very important, however insignificant it may look. I would like to express my special thanks to Prof. Michael Raghunath, who not only taught us basic methods of research as a mentor and teacher but also helped me beyond limits for my PhD completion. This thesis would not have been possible without his supportive lessons throughout my PhD carrier and his special role for my thesis completion. I owe my deepest gratitude to Prof Dietmar W. Hutmacher, who constantly guided me from the beginning until the very end of this PhD journey. I am very grateful to work with such a scientific and fatherly figure. His supportive words through skype web chatting and his magical emails made me feel his presence in each step of this journey even without his physical presence. I would like to express my sincere and overwhelming gratitude to Dr. med. Ulrich Kneser, and Dr. J. Thorsten Schantz for their guidance and invaluable advice imparted throughout this project work. I remain very much indebted to all my supervisors for their magnanimous and unflinching inspiration instilled in me. The knowledge I gained from each of them in different areas of my work will be helpful to dedicate my life to science. i The support and guidance I received from the Lab of Tissue Engineering in NUS and DSO NUSTEP, Singapore, were exemplary and made my research experience memorable. I also take this opportunity to thank Lam Xu Fu Chirstopher, PhD student in Lab. of Tissue Engineering, who guided and helped me in all scaffold related work throughout my research work. I am indebted to him to support right until the end of my thesis work. I express my sincere thanks to Barney for his microCT lessons; Andrew for his works related to Extracel-HP. I am grateful to Clarice, Dr. Gajadhar Bhakta, Evelyne, Anurag, David, Monique, Anand and Dr. Kee Woei for their timely advice and knowledge in every aspect of my PhD work. I am indeed fortunate to have worked alongside such committed and hardworking people. I would like to show my special gratitude to Dr. Sambit Sahoo and Dr. Sampurna Sattar for their moral support as well as scientific inputs in every new experiment. The support for this work by my seniors, especially Dr. Sriram Vedula, Dr Dev Chatterjee, and Dr. Karthik Harve are gratefully acknowledged. I humbly express my sincere gratitude to Dr. Galyna Pryymachuk for her invaluable guidance, and advice imparted during this investigation. I remain very much beholden to her to teach me the microsurgical techniques for the orderly and successful execution of the experiments for the completion of this thesis. I would like to thank Dr Andreas Arkudas for his wise thoughts and kind words of advice at every point in my endeavour without which it would not have been possible to realize the thesis work. My special thanks go to Dr. Elias Polykandroitis for contributing many ideas during the initial phase of my project. I would also like to thank Dr. Oliver Bleizifer, Dr. Saskia Schnabl, and Dr. Justus Bier to be helpful with friendly advices throughout my work. ii I am also grateful to many people who have helped and contributed, in one way or another, to my research project. It has been a great pleasure to have worked with Stefan Fleischer, Katja Schubert, and Dorothee Klummp, who have been indispensable in the smooth completion of the project work. I would like to thank all staff in division of bioengineering, NUS and department of plastic surgery, University Hospital, Erlangen for their official and financial support. Last, but not the least, I would also like to thank my family, especially my parents; my wife, Bagmi; my brothers Bibhu and Prabhu for being very encouraging and supportive throughout my PhD career. July 2009 (Dr. Subha Narayan Rath) National University of Singapore iii List of published work • Leong DT, Abraham MC, Rath SN, Lim TC, Chew FT, Hutmacher DW. Investigating the effects of preinduction on human adipose-derived precursor cells in an athymic rat model. Differentiation. 2006 Dec; 74(9-10):519-29. • Rath SN, Woodruff MA, Susanto E, Haupt LM, Hutmacher DW, Nurcombe V, Cool SM. Sustained release and osteogenic potential of heparan sulfate-doped fibrin glue scaffolds within a rat cranial model. J Mol Histol. 2007 Sep 12. • Rath SN, Cohn D, Hutmacher DW. Comparison of Chondrogenesis in Static and Dynamic Environment Using PCL-PEO Scaffold. Accepted in Journal of Virtual and Physical Prototyping. • Fiegel HC , Pryymachuk G , Rath S , Bleiziffer O , Beier JP , Bruns H , Kluth D , Metzger R , Horch RE , Till H , Kneser U . Fetal Hepatocyte Transplantation in a Vascularized AV-Loop Transplantation Model in the Rat. Journal of cellular and molecular medicine. 2008 May 24. H H H H H H H H H H H H H H H H H H H H H H • Rath SN, Arkudas A, Pryymachuk G, Polykandroitis E, Christopher LXF, Bier JP, Horch RE, Hutmacher DW, Kneser U. Development of a Pre-vascularized 3D Composite ScaffoldHydrogel System Using an Artery-Venous Loop for Tissue Engineering Applications; submitted to Biomaterials. • Rath SN, Pryymachuk G, Bleiziffer OA, Lam CXF, Arkudas A, Ho STB, Bier JP, Horch RE, Hutmacher DW, Kneser U. Hyaluronan-based heparin-incorporated hydrogels for generation of axially vascularized bioartificial bone tissues: in vitro and in vivo evaluation in a PLDLLATCP-PCL-composite system; submitted to Tissue Engineering. iv Table of contents Acknowledgements . i List of published work iv Table of contents v Summary . x List of tables xiii List of figures . xiv List of symbols xvii CHAPTER 1. Introduction 1.1. Background 1.2. Tissue engineering . 1.2.1 Importance of vascularization in tissue engineering 1.2.2 Making a graft vascularized . 1.3. Aims and hypotheses . 1.3.1 Specific aim 1: To establish the scaffold architecture and optimize the artery-venous (A-V) loop model in rat using the scaffold and to standardize the explantation procedure. . 1.3.2 Specific aim 2: To develop and evaluate pre-vascularized 3D composite scaffold-hydrogel systems using an A-V loop for a possible graft. . 1.3.3 Specific aim 3: Application of bone morphogenetic protein-2 (BMP-2) or osteoblasts in the vascularized composite scaffold-modified hyaluronan hydrogel system for development of a vascularized bone graft. . 1.4. Organization of the Thesis . CHAPTER 2. Background to the Research . 10 2.1. Angiogenesis and vascularization 10 2.2. Importance of vascularization in tissue-specific tissue engineering 13 2.2.1 Bone tissue engineering 13 2.2.2 Skin tissue engineering . 15 v 2.2.3 Genito-urinary tissue engineering 16 2.2.4 Limitations of current status of tissue engineering . 16 2.3. Methods of generating vascularized grafts 18 2.3.1 Cell Source . 18 2.3.2 Scaffold Material and design 20 2.3.3 Use of growth factors . 21 2.3.4 Gene therapy for angiogenesis . 22 2.3.5 Extracellular Matrix Components 23 2.4. Surgical approach with artery-venous loop . 25 2.4.1 Cell free method: using body as bioreactor 30 CHAPTER 3. Experiment I: To establish the scaffold architecture and optimize the artery-venous (A-V) loop model in rat using the scaffold and to standardize the explantation procedure. 32 3.1. Introduction 32 3.2. Materials and methods . 35 3.2.1 Scaffold fabrication 35 3.2.2 Porosity and pore interconnectivity calculation . 37 3.2.3 Mechanical testing 38 3.2.4 A-V loop model 39 3.2.5 Explantation 40 3.2.6 Micro CT analysis 40 3.2.7 Histology 41 3.2.8 Statistical analysis 42 3.3. Results 43 3.3.1 Scaffold gross morphology . 43 3.3.2 Porosity and interconnectivity 43 3.3.3 Mechanical testing 44 3.3.4 Explantation 44 3.3.5 Histology 46 vi 3.4. Discussion 46 3.5. Conclusion . 51 CHAPTER 4. Experiment II: Development and characterization of a pre- vascularized 3D composite scaffold-hydrogel system using an artery-venous loop for tissue engineering applications. 53 4.1. Introduction 53 4.2. Materials & methods 56 4.2.1 Scaffold fabrication 56 4.2.2 Experimental design . 57 4.2.3 Hydrogels . 58 4.2.4 Surgical procedures 58 4.2.5 Explantation 59 4.2.6 Micro CT analysis 59 4.2.7 Histology 60 4.2.8 Histomorphometric analysis . 60 4.2.9 Immunohistochemistry . 61 4.2.10 Vascular corrosion cast preparation . 61 4.2.11 Scanning electron microscopy 62 4.2.12 Statistical analysis 62 4.3. Results: 63 4.3.1 Surgery and animals: 63 4.3.2 Micro CT analysis: . 63 4.3.3 Histology & Corrosion casting: 64 4.3.4 Histomorphometry 67 4.3.5 Immunohistochemistry . 69 4.4. Discussion: . 70 4.5. Conclusion: 74 vii CHAPTER 5. Experiment III: Applying bone morphogenetic protein-2 (BMP-2) or osteoblasts in the composite scaffold-modified hyaluronan hydrogel system along with A-V loop for a vascularized bone graft formation. 76 5.1. Introduction 76 5.2. Materials and methods . 78 5.2.1 Scaffold fabrication 78 5.2.2 Osteoblasts isolation and expansion . 78 5.2.3 Osteoblast culture in modified hyaluronan hydrogel . 81 5.2.4 AlamarBlue metabolic assay 82 5.2.5 PicoGreen DNA Quantification Assay . 83 5.2.6 FDA/PI fluorescent staining of osteoblasts 83 5.2.7 Release kinetics of rhBMP2 from hyaluronan hydrogel (Extracel-HP) . 84 5.2.8 Experimental design . 85 5.2.9 Surgical procedures and explantation . 86 5.2.10 RNA isolation and quantitative RT-PCR . 86 5.2.11 Micro CT analysis 87 5.2.12 Histology 88 5.2.13 Histomorphometric analysis . 88 5.2.14 Immunohistochemistry . 88 5.2.15 Corrosion cast technique 88 5.2.16 Statistical analysis 88 5.3. Results 89 5.3.1 Osteoblasts in Hyaluronan hydrogel 89 5.3.2 Release kinetics 91 5.3.3 Surgery and animals . 92 5.3.4 Micro CT analysis 92 5.3.5 Histology 95 5.3.6 Histomorphometry 97 5.3.7 Corrosion casting 98 viii 5.3.8 Immunohistochemistry . 99 5.3.9 Real time PCR 101 5.4. Discussion 103 5.5. Conclusion . 111 CHAPTER 6. CO CLUSIO S A D RECOMME DATIO S . 113 6.1. Conclusions 113 6.2. Recommendations for future research . 116 Reference: . 118 ix Chapter 6. Conclusions and recommendations CHAPTER 6. CO CLUSIO S A D RECOMME DATIO S 6.1. Conclusions Starting from metal alloys, the cells and growth-factor based biological constructs impact a drastic change for bone tissue engineering applications. Although scientists still attempt to understand the basic bone physiology and the role of different transcription factors for bone development, a number of composite biomaterials are practiced for bone fracture and bone loss treatments. The attempts and research publications to mimic the anatomy of bone by using a combination of composite materials, growth factors, mesenchymal stem cells or osteoblasts provide a strong hope for a realistically fulfillment of a suitable tissue engineered product in near future. With the advent of bioreactors and well established differentiation methods for stem cells, a highly suitable bone product in the laboratory seems feasible. However, a number of important issues still need to be addressed. Despite some success, tissue engineered bone products have not become widely in use, indicating some unresolved issues for in vivo use. The autograft from iliac crest continues to be the gold standard in bone loss cases. In this PhD research, a novel ceramic polymer composite biomaterial for bone tissue engineering approach was evaluated, in order to sustain the mechanical load of bone in spite of highly porous structure for effective tissue in-growth. A systematic approach was attempted to develop a vascularized bone graft with PLDLLA-TCPPCL scaffolds and hyaluronan hydrogel along with additives for proper differentiation of the tissue construct. The biomaterial needs to be biologically well compatible for viable bone in-growth. Therefore, the biomaterial with a specific shape and along with 113 Chapter 6. Conclusions and recommendations a hydrogel was evaluated with microsurgically created A-V loop for possible fibrovascular tissue development and new sprouting angiogenesis from the loop. This technique has produced an established A-V loop supplied composite scaffold hydrogel system for further exploration for successful bone tissue engineering application. The experiments conducted were focused on the development of a novel vascularized biomaterial for prolonged survival of cells or growth factors to develop a well-differentiated tissue. In this PhD thesis, The PLDLLA-TCP-PCL scaffold shape was attempted to provide the basic shape of the final construct, while all the extra additives can be helpful for changing the progress towards bone and angiogenesis growth deep into the construct. The experiments show that the mechanical strength and shape will be provided by the PLDLLA-TCP-PCL scaffold; but the A-V loop will provide angiogenesis and make the whole construct viable. The hydrogel will be helpful not only for allowing the ingrowth of angiogenesis but also the additions of growth factors or cells for proper differentiation of the construct. This is biologically more relevant and similar to bone anatomy and function. Furthermore, the physical properties of the scaffold can be changed with its two different parts namely, the ceramic part providing the strength and bioactivity, while the polymer part providing relatively quick degradability and shear stress sustainability. However, the microsurgically constructed A-V loop provides the main nutrition for the survival of the whole construct and later can make it act like a vascularized autograft. Based on the above strategy, the vascularized scaffold hydrogel system can keep the osteoblasts survived. Although a well defined bone was absent histologically, still there is favourable gene expression for bone markers. Assessment of angiogenesis pattern and progression in those two different hydrogels indicated that there were 114 Chapter 6. Conclusions and recommendations significant differences between them. The vascular growth in fibrin glue is related to the quick degradation of the hydrogel in vivo; however, the density and pattern after eight weeks are not comparatively more relative to four weeks. Moreover, the growth in the hyaluronan matrix is slow and progressive in relation to its slow degradation in vivo and after eight-week time point the fibro-vascular growth was very much similar to that of fibrin glue hydrogel. The A-V loop based scaffold has also been shown to be well suited for further exploration of growth factors or cells. In conclusion, this project has devised and demonstrated a method that can precisely engineer a highly vascularized scaffold for bone tissue engineering application, and yet, is versatile enough to be readily adapted into further experiment for different tissue growth by applying extra additives. When we analyze the specific objectives postulated in Chapter 1, the following conclusions were drawn from the experimental results: • A synthetic PLDLLA-TCP-PCL scaffold with mechanical properties similar to a cancellous bone was fabricated and for thorough vascularization of such a biomaterial the artery-venous loop was established and analyzed after Microfil injection and explantation. • The PLDLLA-TCP-PCL scaffold along with two different hydrogels namely, fibrin glue and hyaluronan were evaluated along with the surgically constructed A-V loop for possible support of angiogenesis and the rate of angiogenesis was analyzed with fast degradation of fibrin glue and slow degradation of the modified hyaluronan matrix. • The modified hyaluronan hydrogel sustains implanted osteoblasts well with alkaline phosphatase positive osteoblasts and high percentage of live cells. • The release kinetic of BMP-2 was slow and the degradation of the modified hyaluronan matrix was prolonged with some intact matrix left even after eight weeks. 115 Chapter 6. Conclusions and recommendations • The scaffold hyaluronan hydrogel system can be used to incorporate BMP2 to support the development of extra-osseous bone for possible bone graft application. • Although bone histology is lacking from the construct in the analyzed eightweek time point, the gene expression shows a well-favored tissue construct for bone graft and highly vascularized tissue for implanted cells to be viable. This technique of combining PLDLLA-TCP-PCL scaffold with modified hyaluronan matrix has the potential to be developed into a clinically viable tissue engineered bone construct. 6.2. Recommendations for future research The results in this PhD research showed that using PLDLLA-TCP-PCL scaffold with a suitable dose of BMP-2 can make a useful well-vascularized bone construct to replace the present gold standard iliac crest autograft for bone loss states. However, for clinically useful application of this bone-construct product, further evaluation of a number of aspects will need to be carried out, as recommended below: 1. In this research, degradation of the PLDLLA-TCP-PCL was not observed in vivo in the observed period of weeks at all. But for clinical application the degradation time and kinetics of this defined product must be studied in vivo. 2. In spite of highly satisfactory angiogenesis growth from A-V loop in the observed eight-week time period, a long term study is desirable for demonstration of the permanent nature of the blood vessels as in many research articles it was shown some seemingly permanent blood vessels actually degenerate with time in a longer duration study [62]. It is therefore, recommended that the A-V loop with the scaffold hydrogel system were studied for almost 6-12 months with proper measurement of angiogenesis. 116 Chapter 6. Conclusions and recommendations 3. With the adopted experimental set-up in this thesis, only the osteoblast implanted scaffolds have intense angiogenesis throughout the scaffold. However, even with growth factors, some areas of central parts still lack new blood vessel growth. It may be due to unsuitable porosity of the scaffolds or the short time period of study. These variations need to be tested to have intense angiogenesis similar to natural tissue. 4. In spite of the fact that a higher percentage of cells or a greater amount of growth factors can be active due to supplied A-V loop, there still needs a lag period of few days in which the growth of new blood vessels occurs. We can postulate another hypothesis that the cells or growth factors applied only after complete vascularization will make more active tissue-engineered product compared to the present status. This hypothesis needs to be tested compared to the present product. However, this needs opening of the wound for two times for cell or growth factor implantation. 5. At the end, regardless of the strategy used, the ultimate success or lack of it can only be established when such a construct after proper evaluation will later be implanted in a bone defect site and then the overall healing is evaluated. Once this success is shown, this can be an alternative to the present gold standard of iliac crest autograft for bone loss patients. The ability of this tissue engineered graft to heal a critical-sized bone defect will therefore, need to be explored. 117 Reference Reference: 1. Shi D. Biomaterials and Tissue Engineering: Springer, 2004. 2. 2006 Organ Donation, Transplantation Rates Continue Growth Trend. U.S. Department of Health & Human Services, Health Resources and Services Administration, 2007. Available from: http://newsroom.hrsa.gov/releases/2007/OrganDonationRates2006.htm 3. Langer R, Vacanti J. Tissue engineering. Science 1993;260(5110):920-926. 4. Knight M, Evans G. Tissue engineering: progress and challenges. Plastic and Reconstructive Surgery 2004;114(2):26. 5. Griffith L. Expanding Opportunities. Science 2002;1069210(1009):295. 6. Bueno EM, Glowacki J. Cell-free and cell-based approaches for bone regeneration. Nat Rev Rheumatol 2009 Dec;5(12):685-697. 7. Mansbridge JN. Tissue-engineered skin substitutes in regenerative medicine. Curr Opin Biotechnol 2009 Oct;20(5):563-567. 8. Wood D, Southgate J. Current status of tissue engineering in urology. Curr Opin Urol 2008 Nov;18(6):564-569. 9. Novicoff WM, Manaswi A, Hogan MV, Brubaker SM, Mihalko WM, Saleh KJ. Critical analysis of the evidence for current technologies in bone-healing and repair. J Bone Joint Surg Am 2008 Feb;90 Suppl 1:85-91. 10. Bishop G, Einhorn T. Current and future clinical applications of bone morphogenetic proteins in orthopaedic trauma surgery. International Orthopaedics 2007;31(6):721-727. 11. Tseng SS, Lee MA, Reddi AH. Nonunions and the potential of stem cells in fracture-healing. J Bone Joint Surg Am 2008 Feb;90 Suppl 1:92-98. 12. Giannoudis P, Dinopoulos H, Tsiridis E. Bone substitutes: An update. Injury 2005;36(3S):2027. 13. Kanczler JM, Oreffo RO. Osteogenesis and angiogenesis: the potential for engineering bone. Eur Cell Mater 2008;15:100-114. 14. Baumert H, Simon P, Hekmati M, Fromont G, Levy M, Balaton A, et al. Development of a seeded scaffold in the great omentum: feasibility of an in vivo bioreactor for bladder tissue engineering. Eur Urol 2007;52(3):884-890. 15. Arkudas A, Tjiawi J, Bleiziffer O, Grabinger L, Polykandriotis E, Beier JP, et al. Fibrin gelimmobilized VEGF and bFGF efficiently stimulate angiogenesis in the AV loop model. Mol Med 2007 Sep-Oct;13(9-10):480-487. 16. Bolland BJRF, Kanczler JM, Dunlop DG, Oreffo ROC. Development of in vivo µCT evaluation of neovascularisation in tissue engineered bone constructs. Bone 2008;43(1):195202. 17. Hiltunen MO, Ruuskanen M, Huuskonen J, Mahonen AJ, Ahonen M, Rutanen J, et al. Adenovirus-mediated VEGF-A gene transfer induces bone formation in vivo. Faseb J 2003 Jun;17(9):1147-1149. 118 Reference 18. Kaigler D, Wang Z, Horger K, Mooney DJ, Krebsbach PH. VEGF scaffolds enhance angiogenesis and bone regeneration in irradiated osseous defects. J Bone Miner Res 2006 May;21(5):735-744. 19. Baffour R, Berman J, Garb JL, Rhee SW, Kaufman J, Friedmann P. Enhanced angiogenesis and growth of collaterals by in vivo administration of recombinant basic fibroblast growth factor in a rabbit model of acute lower limb ischemia: dose-response effect of basic fibroblast growth factor. J Vasc Surg 1992 Aug;16(2):181-191. 20. Berse B, Hunt JA, Diegel RJ, Morganelli P, Yeo K, Brown F, et al. Hypoxia augments cytokine (transforming growth factor-beta (TGF-beta) and IL-1)-induced vascular endothelial growth factor secretion by human synovial fibroblasts. Clin Exp Immunol 1999;115(1):176182. 21. Schilephake H. Bone growth factors in maxillofacial skeletal reconstruction. Int J Oral Maxillofac Surg 2002 Oct;31(5):469-484. 22. Komatsu D, Hadjiargyrou M. Activation of the transcription factor HIF-1 and its target genes, VEGF, HO-1, iNOS, during fracture repair. Bone 2004;34(4):680-688. 23. Alajati A, Laib AM, Weber H, Boos AM, Bartol A, Ikenberg K, et al. Spheroid-based engineering of a human vasculature in mice. Nat Methods 2008 May;5(5):439-445. 24. Nomi M, Miyake H, Sugita Y, Fujisawa M, Soker S. Role of growth factors and endothelial cells in therapeutic angiogenesis and tissue engineering. Curr Stem Cell Res Ther 2006 Sep;1(3):333-343. 25. Kung EF, Wang F, Schechner JS. In vivo perfusion of human skin substitutes with microvessels formed by adult circulating endothelial progenitor cells. Dermatol Surg 2008 Feb;34(2):137-146. 26. Zisch AH. Tissue engineering of angiogenesis with autologous endothelial progenitor cells. Curr Opin Biotechnol 2004 Oct;15(5):424-429. 27. Degistirici O, Jager M, Knipper A. Applicability of cord blood-derived unrestricted somatic stem cells in tissue engineering concepts. Cell Prolif 2008 Jun;41(3):421-440. 28. Wenger A, Stahl A, Weber H, Finkenzeller G, Augustin HG, Stark GB, et al. Modulation of in vitro angiogenesis in a three-dimensional spheroidal coculture model for bone tissue engineering. Tissue Eng 2004 Sep-Oct;10(9-10):1536-1547. 29. Dvorak HF. Angiogenesis: update 2005. J Thromb Haemost 2005;3(8):1835-1842. 30. Kneser U, Polykandriotis E, Ohnolz J, Heidner K, Grabinger L, Euler S, et al. Engineering of vascularized transplantable bone tissues: induction of axial vascularization in an osteoconductive matrix using an arteriovenous loop. Tissue Eng 2006 Jul;12(7):1721-1731. 31. Mian R, Morrison WA, Hurley JV, Penington AJ, Romeo R, Tanaka Y, et al. Formation of new tissue from an arteriovenous loop in the absence of added extracellular matrix. Tissue Eng 2000 Dec;6(6):595-603. 32. Tanaka Y, Tsutsumi A, Crowe DM, Tajima S, Morrison WA. Generation of an autologous tissue (matrix) flap by combining an arteriovenous shunt loop with artificial skin in rats: preliminary report. Br J Plast Surg 2000 Jan;53(1):51-57. 33. Hofer SO, Knight KM, Cooper-White JJ, O'Connor AJ, Perera JM, Romeo-Meeuw R, et al. Increasing the volume of vascularized tissue formation in engineered constructs: an experimental study in rats. Plast Reconstr Surg 2003 Mar;111(3):1186-1192; discussion 11931184. 119 Reference 34. Erol Ö, Spira M. New Capillary Bed Formation with a Surgically Constructed Arteriovenous Fistula. Plastic and Reconstructive Surgery 1980;66(1):109. 35. Rafii S, Meeus S, Dias S, Hattori K, Heissig B, Shmelkov S, et al. Contribution of marrowderived progenitors to vascular and cardiac regeneration. Seminars in Cell and Developmental Biology 2002;13(1):61-67. 36. Pettersson A, Nagy JA, Brown LF, Sundberg C, Morgan E, Jungles S, et al. Heterogeneity of the Angiogenic Response Induced in Different Normal Adult Tissues by Vascular Permeability Factor/Vascular Endothelial Growth Factor. Lab Invest 2000;80:99-115. 37. Theile DR, Kane AJ, Romeo R, Mitchell G, Crowe D, Stewart AG, et al. A model of bridging angiogenesis in the rat. Br J Plast Surg 1998 Apr;51(3):243-249. 38. Birch J, BråRnemark P. The Vascularization of a Free Full Thickness Skin Graft: I. A Vital Microscopic Study. Scandinavian Journal of Plastic and Reconstructive Surgery and Hand Surgery 1969;3(1):1-10. 39. Ausprunk D, Knighton D, Folkman J. Differentiation of vascular endothelium in the chick chorioallantois: a structural and autoradiographic study. Dev Biol 1974;38(2):237-248. 40. Ryu S, Albert D. Evaluation of tumor angiogenesis factor with the rabbit cornea model. Investigative Ophthalmology & Visual Science 1979;18(8):831-841. 41. Dellian M, Witwer B, Salehi H, Yuan F, Jain R. Quantitation and physiological characterization of angiogenic vessels in mice: effect of basic fibroblast growth factor, vascular endothelial growth factor/vascular permeability factor, and host microenvironment. American Journal of Pathology 1996;149(1):59-71. 42. Polykandriotis E, Arkudas A, Horch RE, Stu?rzl M, Kneser U. Autonomously vascularized cellular constructs in tissue engineering: Opening a new perspective for biomedical science. Journal of Cellular and Molecular Medicine 2007;11(1):6-20. 43. Cronin KJ, Messina A, Knight KR, Cooper-White JJ, Stevens GW, Penington AJ, et al. New murine model of spontaneous autologous tissue engineering, combining an arteriovenous pedicle with matrix materials. Plast Reconstr Surg 2004 Jan;113(1):260-269. 44. Bran GM, Stern-Straeter J, Hormann K, Riedel F, Goessler UR. Apoptosis in bone for tissue engineering. Arch Med Res 2008 Jul;39(5):467-482. 45. Terheyden H, Warnke P, Dunsche A, Jepsen S, Brenner W, Palmie S, et al. Mandibular reconstruction with prefabricated vascularized bone grafts using recombinant human osteogenic protein-1: an experimental study in miniature pigs. Part II: transplantation. Int J Oral Maxillofac Surg 2001 Dec;30(6):469-478. 46. Egaña J, Danner S, Kremer M, Rapoport D, Lohmeyer J, Dye J, et al. The use of glandularderived stem cells to improve vascularization in scaffold-mediated dermal regeneration. Biomaterials 2009;30(30):5918-5926. 47. Atala A. Regenerative medicine and tissue engineering in urology. Urol Clin North Am 2009 May;36(2):199-209. 48. Theile DRB, Kane AJ, Romeo R, Mitchell G, Crowe D, Stewart AG, et al. A model of bridging angiogenesis in the rat. British Journal of Plastic Surgery 1998;51(3):243-249. 49. Pribaz JJ, Fine NA. Prelamination: defining the prefabricated flap--a case report and review. Microsurgery 1994;15(9):618-623. 120 Reference 50. Gartsman GM, Weiland AJ, Moore JR, Randolph MA. Blood vessel implantation into ischemic bone. J Reconstr Microsurg 1985 Jan;1(3):215-222. 51. Hori Y, Tamai S, Okuda H, Sakamoto H, Takita T, Masuhara K. Blood vessel transplantation to bone. J Hand Surg [Am] 1979 Jan;4(1):23-33. 52. Men B, Ma C, Li Z. [Revascularization of isolated bone in dogs]. Zhonghua Wai Ke Za Zhi 1997 May;35(5):307-309. 53. Warnke PH, Springer ING, Wiltfang J, Acil Y, Eufinger H, Wehmller M, et al. Growth and transplantation of a custom vascularised bone graft in a man. The Lancet;364(9436):766-770. 54. Warnke PH, Wiltfang J, Springer I, Acil Y, Bolte H, Kosmahl M, et al. Man as living bioreactor: Fate of an exogenously prepared customized tissue-engineered mandible. Biomaterials 2006;27(17):3163-3167. 55. Gill DR, Ireland DC, Hurley JV, Morrison WA. The prefabrication of a bone graft in a rat model. J Hand Surg [Am] 1998 Mar;23(2):312-321. 56. Adams JC. Outline of fractures, including joint injuries. 11th ed: Churchill Livingstone, 1999. 57. Taylor GI. The current status of free vascularized bone grafts. Clin Plast Surg 1983 Jan;10(1):185-209. 58. Alam MI, Asahina I, Seto I, Oda M, Enomoto S. Prefabrication of vascularized bone flap induced by recombinant human bone morphogenetic protein (rhBMP-2). Int J Oral Maxillofac Surg 2003 Oct;32(5):508-514. 59. Wu X, Rabkin-Aikawa E, Guleserian KJ, Perry TE, Masuda Y, Sutherland FWH, et al. Tissue-engineered microvessels on three-dimensional biodegradable scaffolds using human endothelial progenitor cells. American Journal of Physiology- Heart and Circulatory Physiology 2004;287(2):480-487. 60. Schechner J, Nath A, Zheng L, Kluger M, Hughes C, Sierra-Honigmann M, et al. In vivo formation of complex microvessels lined by human endothelial cells in an immunodeficient mouse. Proceedings of the National Academy of Sciences of the United States of America 2000;97(16):9191. 61. Nör JE, Peters MC, Christensen JB, Sutorik MM, Linn S, Khan MK, et al. Engineering and Characterization of Functional Human Microvessels in Immunodeficient Mice. Lab Invest 2001;81:453-463. 62. Koike N, Fukumura D, Gralla O, Au P, Schechner JS, Jain RK. Tissue engineering: creation of long-lasting blood vessels. Nature 2004;428(6979):138-139. 63. Muschler G, Nakamoto C, Griffith L. Engineering Principles of Clinical Cell-Based Tissue Engineering. The Journal of Bone and Joint Surgery 2004;86(7):1541-1558. 64. Karageorgiou V, Kaplan D. Porosity of 3D biomaterial scaffolds and osteogenesis. Biomaterials 2005;26(27):5474-5491. 65. Logeart-Avramoglou D, Anagnostou F, Bizios R, Petite H. Engineering bone: challenges and obstacles. Journal of Cellular and Molecular Medicine 2005;9(1):72-84. 66. RIPAMONTI U, MA S, REDDI A. The critical role of geometry of porous hydroxyapatite delivery system in induction of bone by osteogenin, a one morphological protein. Matrix(Stuttgart) 1992;12(3):202-212. 121 Reference 67. Tsuruga E, Takita H, Itoh H, Wakisaka Y, Kuboki Y. Pore Size of Porous Hydroxyapatite as the Cell-Substratum Controls BMP-Induced Osteogenesis. Journal of Biochemistry 1997;121(2):317. 68. Ferrara N, Houck KA, Jakeman LB, Winer J, Leung DW. The vascular endothelial growth factor family of polypeptides. J Cell Biochem 1991 Nov;47(3):211-218. 69. Soker S, Svahn CM, Neufeld G. Vascular endothelial growth factor is inactivated by binding to alpha 2-macroglobulin and the binding is inhibited by heparin. J Biol Chem 1993 Apr 15;268(11):7685-7691. 70. Brown LF, Detmar M, Claffey K, Nagy JA, Feng D, Dvorak AM, et al. Vascular permeability factor/vascular endothelial growth factor: a multifunctional angiogenic cytokine. EXS 1997;79:233-269. 71. Yancopoulos GD, Davis S, Gale NW, Rudge JS, Wiegand SJ, Holash J. Vascular-specific growth factors and blood vessel formation. Nature 2000;407(6801):242-248. 72. Bauters C, Asahara T, Zheng LP, Takeshita S, Bunting S, Ferrara N, et al. Site-specific therapeutic angiogenesis after systemic administration of vascular endothelial growth factor. Journal of Vascular Surgery 1995;21(2):314-325. 73. Pu LQ, Sniderman AD, Brassard R, Lachapelle KJ, Graham AM, Lisbona R, et al. Enhanced revascularization of the ischemic limb by angiogenic therapy. Circulation 1993 Jul;88(1):208215. 74. Zachary I, Gliki G. Signaling transduction mechanisms mediating biological actions of the vascular endothelial growth factor family. Cardiovascular Research 2001;49(3):568-581. 75. Shintani S, Li C, Ishikawa T, Mihara M, Nakashiro K-i, Hamakawa H. Expression of vascular endothelial growth factor A, B, C, and D in oral squamous cell carcinoma. Oral Oncology 2004;40(1):13-20. 76. Peters MC, Polverini PJ, Mooney DJ. Engineering vascular networks in porous polymer matrices. J Biomed Mater Res 2002;60(4):668-678. 77. Hidaka C, Ibarra C, Hannafin JA, Torzilli PA, Quitoriano M, Jen SS, et al. Formation of vascularized meniscal tissue by combining gene therapy with tissue engineering. Tissue Eng 2002 Feb;8(1):93-105. 78. González Á. 2007 Available from: http://seattletimes.nwsource.com/html/businesstechnology/2003809552_tgen28.html 79. Luginbuehl V, Meinel L, Merkle HP, Gander B. Localized delivery of growth factors for bone repair. European Journal of Pharmaceutics and Biopharmaceutics 2004;58(2):197-208. 80. Elisseeff J, Puleo C, Yang F, Sharma B. Advances in skeletal tissue engineering with hydrogels. Orthodontics and craniofacial research 2005;8(3):150. 81. Reddi AH. Bone morphogenetic proteins: an unconventional approach to isolation of first mammalian morphogens. Cytokine Growth Factor Rev 1997;8(1):11-20. 82. Colton CK. Implantable biohybrid artificial organs. Cell Transplant 1995;4(4):415-436. 83. Kneser U, Schaefer DJ, Polykandriotis E, Horch RE. Tissue engineering of bone: the reconstructive surgeon's point of view. J Cell Mol Med 2006 Jan-Mar;10(1):7-19. 122 Reference 84. Lokmic Z, Stillaert F, Morrison WA, Thompson EW, Mitchell GM. An arteriovenous loop in a protected space generates a permanent, highly vascular, tissue-engineered construct. FASEB J 2007 Feb;21(2):511-522. 85. Abbase EA, Shenaq SM, Spira M, el-Falaky MH. Prefabricated flaps: experimental and clinical review. Plast Reconstr Surg 1995 Oct;96(5):1218-1225. 86. Jilka R, Weinstein R, Bellido T, Parfitt A, Manolagas S. Osteoblast Programmed Cell Death (Apoptosis): Modulation by Growth Factors and Cytokines. Journal of Bone and Mineral Research 1998;13(5):793-802. 87. Levine JP, Bradley J, Turk AE, Ricci JL, Benedict JJ, Steiner G, et al. Bone morphogenetic protein promotes vascularization and osteoinduction in preformed hydroxyapatite in the rabbit. Ann Plast Surg 1997 Aug;39(2):158-168. 88. Seal B, Otero T, Panitch A. Polymeric biomaterials for tissue and organ regeneration. Materials Science & Engineering R 2001;34(4-5):147-230. 89. Schmidmaier G, Wildemann B, Bail H, Lucke M, Fuchs T, Stemberger A, et al. Local application of growth factors (insulin-like growth factor-1 and transforming growth factor-ß1) from a biodegradable poly (d, l-lactide) coating of osteosynthetic implants accelerates fracture healing in rats. Bone 2001;28(4):341-350. 90. Pitt C, Gratzl M, Kimmel G, Surles J, Schindler A. Aliphatic polyesters II. The degradation of poly (DL-lactide), poly (epsilon-caprolactone), and their copolymers in vivo. Biomaterials 1981;2(4):215-220. 91. Martin C, Winet H, Bao J. Acidity near eroding polylactide-polyglycolide in vitro and in vivo in rabbit tibial bone chambers. Biomaterials 1996;17(24):2373-2380. 92. Niiranen H, Pyhalto T, Rokkanen P, Kellomaki M, Tormala P. In Vitro and In Vivo Behavior of Self-Reinforced Bioabsorbable Polymer and Self-Reinforced Bioabsorbable Polymer/Bioactive Glass Composites. Journal of Biomedical Materials Research Part A 2004;69(4):699-708. 93. Dunn A, Campbell P, Marra K. The influence of polymer blend composition on the degradation of polymer/hydroxyapatite biomaterials. Journal of Materials Science: Materials in Medicine 2001;12(8):673-677. 94. Heidemann W, Jeschkeit S, Ruffieux K, Fischer J, Wagner M, Krüger G, et al. Degradation of poly (d, l) lactide implants with or without addition of calciumphosphates in vivo. Biomaterials 2001;22(17):2371-2381. 95. Hammerle C, Olah A, Schmid J, Fluckiger L, Gogolewski S, Winkler J, et al. The biological effect of natural bone mineral on bone neoformation on the rabbit skull. Clin Oral Implants Res 1997;8(3):198-207. 96. Kim H, Knowles J, Kim H. Hydroxyapatite/poly (e-caprolactone) composite coatings on hydroxyapatite porous bone scaffold for drug delivery. Biomaterials 2004;25(7-8):1279-1287. 97. Day R, Boccaccini A, Shurey S, Roether J, Forbes A, Hench L, et al. Assessment of polyglycolic acid mesh and bioactive glass for soft-tissue engineering scaffolds. Biomaterials 2004;25(27):5857-5866. 98. Gatti A, Valdre G, Andersson O. Analysis of the in vivo reactions of a bioactive glass in soft and hard tissue. Biomaterials 1994;15(3):208-212. 99. Boccaccini A, Maquet V. Bioresorbable and bioactive polymer/Bioglass® composites with tailored pore structure for tissue engineering applications. Composites Science and Technology 2003;63(16):2417-2429. 123 Reference 100. Rezwan K, Chen QZ, Blaker JJ, Boccaccini AR. Biodegradable and bioactive porous polymer/inorganic composite scaffolds for bone tissue engineering. Biomaterials 2006 Jun;27(18):3413-3431. 101. Tsuruga E, Takita H, Itoh H, Wakisaka Y, Kuboki Y. Pore size of porous hydroxyapatite as the cell-substratum controls BMP-induced osteogenesis. J Biochem 1997 Feb;121(2):317-324. 102. Hutmacher DW. Scaffold design and fabrication technologies for engineering tissues--state of the art and future perspectives. J Biomater Sci Polym Ed 2001;12(1):107-124. 103. Crump SS, inventor; Stratasys, Inc, assignee. Apparatus and Method for Creating Threedimensional Objects. United States, 1992. 104. Hutmacher DW, Schantz JT, Lam CX, Tan KC, Lim TC. State of the art and future directions of scaffold-based bone engineering from a biomaterials perspective. J Tissue Eng Regen Med 2007 Jul-Aug;1(4):245-260. 105. Zein I, Hutmacher DW, Tan KC, Teoh SH. Fused deposition modeling of novel scaffold architectures for tissue engineering applications. Biomaterials 2002 Feb;23(4):1169-1185. 106. Hutmacher DW, Schantz T, Zein I, Ng KW, Teoh SH, Tan KC. Mechanical properties and cell cultural response of polycaprolactone scaffolds designed and fabricated via fused deposition modeling. J Biomed Mater Res 2001 May;55(2):203-216. 107. Engelke K, Karolczak M, Lutz A, Seibert U, Schaller S, Kalender W. Micro-CT. Technology and application for assessing bone structure. Radiologe 1999;39(3):203-212. 108. Monsivais J. Microvascular grafts: effect of diameter discrepancy on patency rates. Microsurgery 1990;11(4):285-287. 109. Athanasiou KA, Zhu C, Lanctot DR, Agrawal CM, Wang X. Fundamentals of biomechanics in tissue engineering of bone. Tissue Eng 2000 Aug;6(4):361-381. 110. Hutmacher DW. Scaffolds in tissue engineering bone and cartilage. Biomaterials 2000 Dec;21(24):2529-2543. 111. Gibson LJ, Ashby MF. Cellular Solids: Structure and Properties 2nd ed: Cambridge University Press, 1997. 112. Lam CXF, Teoh SH, Hutmacher DW. Comparison of Degradation of PCL & PCL-TCP scaffolds in Alkaline Medium. Polym Int 2007;56(6):718-728. 113. Thomson RC, Yaszemski MJ, Powers JM, Mikos AG. Fabrication of Biodegradable Polymer Scaffolds to Engineer Trabecular Bone. Journal of Biomaterials Science-Polymer Edition 1995;7(1):23-38. 114. De Groot JH, Kuijper HW, Pennings AJ. A novel method for fabrication of biodegradable scaffolds with high compression moduli. Journal of Materials Science-Materials in Medicine 1997 Nov;8(11):707-712. 115. Slivka MA, Leatherbury NC, Kieswetter K, Niederauer GG. Porous, resorbable, fiberreinforced scaffolds tailored for articular cartilage repair. Tissue Engineering 2001 Dec;7(6):767-780. 116. Lohmann CH, Schwartz Z, Niederauer GG, Carnes DL, Jr., Dean DD, Boyan BD. Pretreatment with platelet derived growth factor-BB modulates the ability of costochondral resting zone chondrocytes incorporated into PLA/PGA scaffolds to form new cartilage in vivo. Biomaterials 2000 Jan;21(1):49-61. 124 Reference 117. Sung HJ, Meredith C, Johnson C, Galis ZS. The effect of scaffold degradation rate on threedimensional cell growth and angiogenesis. Biomaterials 2004 Nov;25(26):5735-5742. 118. Azzarello J, Ihnat MA, Kropp BP, Warnke LA, Lin HK. Assessment of angiogenic properties of biomaterials using the chicken embryo chorioallantoic membrane assay. Biomed Mater 2007;2:55–61. 119. Endres M, Hutmacher DW, Salgado AJ, Kaps C, Ringe J, Reis RL, et al. Osteogenic induction of human bone marrow-derived mesenchymal progenitor cells in novel synthetic polymerhydrogel matrices. Tissue Eng 2003 Aug;9(4):689-702. 120. Yamaoka H, Asato H, Ogasawara T, Nishizawa S, Takahashi T, Nakatsuka T, et al. Cartilage tissue engineering using human auricular chondrocytes embedded in different hydrogel materials. J Biomed Mater Res A 2006 Jul;78(1):1-11. 121. Terada S, Yoshimoto H, Fuchs JR, Sato M, Pomerantseva I, Selig MK, et al. Hydrogel optimization for cultured elastic chondrocytes seeded onto a polyglycolic acid scaffold. J Biomed Mater Res A 2005 Dec 15;75(4):907-916. 122. Marijnissen WJ, van Osch GJ, Aigner J, van der Veen SW, Hollander AP, Verwoerd-Verhoef HL, et al. Alginate as a chondrocyte-delivery substance in combination with a non-woven scaffold for cartilage tissue engineering. Biomaterials 2002 Mar;23(6):1511-1517. 123. Heywood HK, Sembi PK, Lee DA, Bader DL. Cellular utilization determines viability and matrix distribution profiles in chondrocyte-seeded alginate constructs. Tissue Eng 2004 SepOct;10(9-10):1467-1479. 124. Nixon AJ, Hendrickson DA, Lust G, Grande DA. Chondrocyte laden fibrin polymers effectively resurface large articular cartilage defects in horses. 38th Annual Meeting, Orthopaedic Research Society, Washington, DC, Orthopaedic Transacations 1992. 125. Hendrickson DA, Nixon AJ, Grande DA, Todhunter RJ, Minor RM, Erb H, et al. Chondrocyte-fibrin matrix transplants for resurfacing extensive articular cartilage defects. J Orthop Res 1994 Jul;12(4):485-497. 126. Bonaventure J, Kadhom N, Cohen-Solal L, Ng KH, Bourguignon J, Lasselin C, et al. Reexpression of cartilage-specific genes by dedifferentiated human articular chondrocytes cultured in alginate beads. Exp Cell Res 1994 May;212(1):97-104. 127. Riley CM, Fuegy PW, Firpo MA, Shu XZ, Prestwich GD, Peattie RA. Stimulation of in vivo angiogenesis using dual growth factor-loaded crosslinked glycosaminoglycan hydrogels. Biomaterials 2006 Dec;27(35):5935-5943. 128. Pike DB, Cai S, Pomraning KR, Firpo MA, Fisher RJ, Shu XZ, 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 Oct;27(30):5242-5251. 129. Cassell OC, Morrison WA, Messina A, Penington AJ, Thompson EW, Stevens GW, et al. The influence of extracellular matrix on the generation of vascularized, engineered, transplantable tissue. Ann N Y Acad Sci 2001 Nov;944:429-442. 130. Tanaka Y, Sung KC, Fumimoto M, Tsutsumi A, Kondo S, Hinohara Y, et al. Prefabricated engineered skin flap using an arteriovenous vascular bundle as a vascular carrier in rabbits. Plast Reconstr Surg 2006 May;117(6):1860-1875. 131. Kneser U, Stangenberg L, Ohnolz J, Buettner O, Stern-Straeter J, Mo?best D, et al. Evaluation of processed bovine cancellous bone matrix seeded with syngenic osteoblasts in a critical size calvarial defect rat model. Journal of Cellular and Molecular Medicine 2006;10(3):695-707. 125 Reference 132. Le Nihouannen D, Guehennec LL, Rouillon T, Pilet P, Bilban M, Layrolle P, et al. Microarchitecture of calcium phosphate granules and fibrin glue composites for bone tissue engineering. Biomaterials 2006;27(13):2716-2722. 133. Jeon O, Ryu SH, Chung JH, Kim BS. Control of basic fibroblast growth factor release from fibrin gel with heparin and concentrations of fibrinogen and thrombin. Journal of Controlled Release 2005;105(3):249-259. 134. Toole BP, Hascall VC. Hyaluronan and tumor growth. Am J Pathol 2002 Sep;161(3):745-747. 135. Rooney P, Kumar S, Ponting J, Wang M. The role of hyaluronan in tumour neovascularization (review). Int J Cancer 1995 Mar 3;60(5):632-636. 136. Shu XZ, Liu Y, Palumbo FS, Luo Y. In situ crosslinkable hyaluronan hydrogels for tissue engineering. Biomaterials 2004;25(7):1339-1348. 137. Cai S, Liu Y, Zheng Shu X, Prestwich G. Injectable glycosaminoglycan hydrogels for controlled release of human basic fibroblast growth factor. Biomaterials 2005;26(30):60546067. 138. Lametschwandtner A, Lametschwandtner U, Weiger T. Scanning electron microscopy of vascular corrosion casts--technique and applications: updated review. Scanning Microsc 1990 Dec;4(4):889-940; discussion 941. 139. Ghajar CM, George SC, Putnam AJ. Matrix metalloproteinase control of capillary morphogenesis. Crit Rev Eukaryot Gene Expr 2008;18(3):251-278. 140. Tonnesen M, Feng X, Clark R. Angiogenesis in Wound Healing. Journal of Investigative Dermatology Symposium Proceedings 2000;5:40-46. 141. Fiegel H, Pryymachuk G, Rath S, Bleiziffer O, Beier J, Bruns H, et al. Fetal Hepatocyte Transplantation in a Vascularized AV-Loop Transplantation Model in the Rat. Journal of Cellular and Molecular Medicine 2008;14(1-2):267–274. 142. Cai S, Liu Y, Zheng Shu X, Prestwich GD. Injectable glycosaminoglycan hydrogels for controlled release of human basic fibroblast growth factor. Biomaterials 2005;26(30):60546067. 143. Patel VV, Zhao L, Wong P, Pradhan BB, Bae HW, Kanim L, et al. An in vitro and in vivo analysis of fibrin glue use to control bone morphogenetic protein diffusion and bone morphogenetic protein–stimulated bone growth. The Spine Journal 2006;6(4):397-403. 144. Inoda H, Yamamoto G, Hattori T. rh-BMP2-induced ectopic bone for grafting critical size defects: a preliminary histological evaluation in rat calvariae. International Journal of Oral and Maxillofacial Surgery 2007;36(1):39-44. 145. Alsberg E, Anderson KW, Albeiruti A, Franceschi RT, Mooney DJ. Cell-interactive alginate hydrogels for bone tissue engineering. J Dent Res 2001 Nov;80(11):2025-2029. 146. Augst AD, Kong HJ, Mooney DJ. Alginate hydrogels as biomaterials. Macromol Biosci 2006 Aug 7;6(8):623-633. 147. Kruyt MC, Persson C, Johansson G, Dhert WJ, de Bruijn JD. Towards injectable cell-based tissue-engineered bone: the effect of different calcium phosphate microparticles and preculturing. Tissue Eng 2006 Feb;12(2):309-317. 148. Schantz JT, Brandwood A, Hutmacher DW, Khor HL, Bittner K. Osteogenic differentiation of mesenchymal progenitor cells in computer designed fibrin-polymer-ceramic scaffolds manufactured by fused deposition modeling. J Mater Sci Mater Med 2005 Sep;16(9):807-819. 126 Reference 149. Jeppsson C, Bostrom M, Aspenberg P. Intraosseous BMP implants in rabbits. Inhibitory effect on bone formation. Acta Orthop Scand 1999 Feb;70(1):77-83. 150. Aspenberg P, Turek T. BMP-2 for intramuscular bone induction: effect in squirrel monkeys is dependent on implantation site. Acta Orthop Scand 1996 Feb;67(1):3-6. 151. Leong DT, Gupta A, Bai HF, Wan G, Yoong LF, Too HP, et al. Absolute quantification of gene expression in biomaterials research using real-time PCR. Biomaterials 2007;28(2):203210. 152. Kakudo N, Kusumoto K, Wang YB, Iguchi Y, Ogawa Y. Immunolocalization of vascular endothelial growth factor on intramuscular ectopic osteoinduction by bone morphogenetic protein-2. Life Sci 2006 Oct 4;79(19):1847-1855. 153. Adams JR, Sander G, Byers S. Expression of hyaluronan synthases and hyaluronidases in the MG63 osteoblast cell line. Matrix Biol 2006 Jan;25(1):40-46. 154. Mazumdar J, Dondeti V, Simon M. Hypoxia-inducible factors in stem cells and cancer. Journal of Cellular and Molecular Medicine 2009;13(11-12):4319-4328. 155. Talwar R, Di Silvio L, Hughes FJ, King GN. Effects of carrier release kinetics on bone morphogenetic protein-2-induced periodontal regeneration in vivo. J Clin Periodontol 2001 Apr;28(4):340-347. 156. Seeherman H, Li R, Bouxsein M, Kim H, Li X, Smith-Adaline E, et al. rhBMP-2/calcium phosphate matrix accelerates osteotomy-site healing in a nonhuman primate model at multiple treatment times and concentrations. The Journal of Bone and Joint Surgery 2006;88(1):144. 157. Rothstein S, Federspiel W, Little S. A unified mathematical model for the prediction of controlled release from surface and bulk eroding polymer matrices. Biomaterials 2009;30(8):1657-1664. 158. Jeon O, Song S, Kang S, Putnam A, Kim B. Enhancement of ectopic bone formation by bone morphogenetic protein-2 released from a heparin-conjugated poly (l-lactic-co-glycolic acid) scaffold. Biomaterials 2007;28(17):2763-2771. 159. Maegawa N, Kawamura K, Hirose M, Yajima H, Takakura Y, Ohgushi H. Enhancement of osteoblastic differentiation of mesenchymal stromal cells cultured by selective combination of bone morphogenetic protein-2 (BMP-2) and fibroblast growth factor-2 (FGF-2). Journal of Tissue Engineering and Regenerative Medicine 2007;1(4):306-313. 160. Noel D, Gazit D, Bouquet C, Apparailly F, Bony C, Plence P, et al. Short-term BMP-2 expression is sufficient for in vivo osteochondral differentiation of mesenchymal stem cells. AlphaMed Press, 2004. p. 74-85. 161. Lecanda F, Avioli LV, Cheng SL. Regulation of bone matrix protein expression and induction of differentiation of human osteoblasts and human bone marrow stromal cells by bone morphogenetic protein-2. J Cell Biochem 1997 Dec 1;67(3):386-396. 162. Murakami T, Saito A, Hino S, Kondo S, Kanemoto S, Chihara K, et al. Signalling mediated by the endoplasmic reticulum stress transducer OASIS is involved in bone formation. Nat Cell Biol 2009 Oct;11(10):1205-1211. 163. Suzuki Y, Montagne K, Nishihara A, Watabe T, Miyazono K. BMPs promote proliferation and migration of endothelial cells via stimulation of VEGF-A/VEGFR2 and angiopoietin1/Tie2 signalling. J Biochem 2008 Feb;143(2):199-206. 164. Golden J, Jones A, Bucholz R, Bosse M, Lyon T, Webb L, et al. Recombinant human BMP-2 and allograft compared with autogenous bone graft for reconstruction of diaphyseal tibial fractures with cortical defects. The Journal of Bone and Joint Surgery 2008;90(5):1168. 127 Reference 165. Lamoureux F, Baud'huin M, Duplomb L, Heymann D, Redini F. Proteoglycans: key partners in bone cell biology. BioEssays 2007;29(8). 166. Reddi A. Role of morphogenetic proteins in skeletal tissue engineering and regeneration. Nature biotechnology 1998;16(3):247-252. 167. Terheyden H, Menzel C, Wang H, Springer IN, Rueger DR, Acil Y. Prefabrication of vascularized bone grafts using recombinant human osteogenic protein-1--part 3: dosage of rhOP-1, the use of external and internal scaffolds. Int J Oral Maxillofac Surg 2004 Mar;33(2):164-172. 128 [...]... surgery are based upon the principle of replacing defective tissues with viable, functional alternatives The two main types of grafts currently in use are autografts and allografts An autograft or autogenous graft is a section of tissue taken from the patient's own body, whereas an allograft is taken from a cadaver In spite of many available treatment modalities for tissue loss, currently, the gold standard... flaps for skin; because the related blood vessels are constant and reasonably suitable for micro surgical manipulations [57] The advantage of vascularized graft is that it can effectively resist infection, can repair the loss of large segmental defects even in a diseased bed, and it does not undergo graft failure [58] In tissue engineering field, many ways are manipulated to make the graft vascularized. .. maintain, or improve tissue functions [3] In the following time, there were a lot of changes in research from the original views given by them to fabricate tissue engineered substances Although all tissues were probable candidates of tissue engineering approach, few tissues such as skin, cartilage, and bone are in advanced stage because of their potential need and relative ease of application 1 Chapter... wound healing in adults [29] The overall rate of vascular in-growth can be stimulated more, if the feeding parent vessels passed through the graft Many researchers have accomplished this by making an artery-venous (A- V) loop to supply blood to the graft [15, 30-33] In most of the cases, a contralateral artery/vein graft is made anastomosis between an artery and a vein for an A- V loop which can readily... make a viable graft Optimization of the interplay of cells and growth factors in the scaffolds might eventually allow generation of different axially vascularized grafts for application in reconstructive surgery This research project makes a promising approach for a vascularized graft for further exploration xii List of tables Table 3-1: The individual components of the composite scaffold and their... from an accident, bone transplants may provide an appropriate solution When there is a necessity of bone transplantation, two options are there: bone from the patient himself may be utilized, which is called autograft or donor bone tissue may be arranged in different forms Although autografts are still considered the best graft, there are limitations to its supply and associated morbidity to already... Numerous studies have been published in an attempt of 17 Chapter 2 Background to the research vascularization of tissues with pedicle graft with varying degrees of success [50-52] At the same time studies are being carried out to test the pre-vascularization of implant before it can be actually used as a transplant in the defect site [53-55] 2.3 Methods of generating vascularized grafts A free graft does not... especially for the central portions of the graft Keeping this view as the main target, a number of research approaches were directed to make a graft viable The cells and factors which are necessary for development of a normal vasculature during embryonic development are recapitulated during situations of neoangiogenesis in adults A number of factors involved in neoangiogenesis can be used such as Vascular... shorter healing time and a graft which is resistance to infection and extrusion This method is called pre-vascularization, which is a process of making neovascularization in tissues by implanting a vascular pedicle into them that can later be transferred to defect site as a graft by micro vascular anastomosis [49] This method has long been applied to skin and other soft tissues in plastic and reconstructive... survival of the graft is dependent upon an intact vascular supply, without which the graft functionality is hampered [13] A bone loss site may be treated by direct application of autografts or osteogenic growth factors for bone formation However, in many cases the local site is compromised by huge bone mass loss, scarring, or irradiation for cancer therapy, without having any healthy tissue If we apply . of a vascularized bone graft. 8 1.4. Organization of the Thesis 9 CHAPTER 2. Background to the Research 10 2.1. Angiogenesis and vascularization 10 2.2. Importance of vascularization in tissue- specific. eventually allow generation of different axially vascularized grafts for application in reconstructive surgery. This research project makes a promising approach for a vascularized graft for. bone are in advanced stage because of their potential need and relative ease of application. Chapter 1. Introduction 2 1.2. Tissue engineering The acute shortage of human tissues and organs

Ngày đăng: 14/09/2015, 08:48

Từ khóa liên quan

Tài liệu cùng người dùng

Tài liệu liên quan