人脂肪干细胞体内外构建组织工程软骨的实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
目的:探讨在体内外环境下,种子细胞、支架材料与环境因素之间的相互作用;明确组织工程软骨对关节软骨损伤模型的治疗作用。
     方法:从人脂肪抽吸物中分离、纯化间充质干细胞,进行体外培养、鉴定;对细胞进行成软骨诱导分化,与海藻酸钠钙凝胶复合,在体外及裸鼠皮下和关节内构建组织工程软骨,观察组织工程软骨对损伤软骨的修复作用及种子细胞、支架材料、环境因素三者之间的相互作用。
     结果:
     1.从人脂肪组织中可以得到大量间充质干细胞。
     2.ADMSCs可成软骨诱导,与海藻酸钠钙凝胶复合后,在微球内分布均匀,细胞呈球形,大部分细胞存活。
     3.组织工程软骨促进裸鼠关节软骨损伤修复,新生软骨与正常软骨的细胞形态及基质成分接近;可见软骨下骨结构形成。
     结论:
     ADMSCs是良好的软骨组织工程种子细胞来源。ADMSCs与海藻酸钠钙凝胶具有良好的组织相容性。经体外成软骨诱导的ADMSCs复合海藻酸钠后,在裸鼠皮下和受损关节内可以保持软骨细胞表型;可以促进其关节软骨损伤的修复。
The defects of articular cartilage induced by injuries or diseases are commonly encountered in hand surgery. Due to its intrinsic properties that lack of vascular supply and the low metabolic activity of chondrocytes, articular cartilage exhibits a limited capacity for intrinsic repair. Thus, caitilage defects, without surgical intervention will either remain injured or progressively worsen with time. Pieces of articular cartilage may become elevated flaps and irritate the synovial lining of the arthrosis causing swelling (effusion) and mechanical symptoms of catching. If the flap of cartilage becomes detached, it can become a loose body, causing locking of the arthrosis so that it does not bend or straighten fully. Surgical techniques and options have focused on the healing of articular cartilage defects. Many of these strategies appear to satisfactory in some cases and can decrease pain and morbidity in the short term, but not effectively sa a long term. Therefore defects of articular cartilage continues to present amajor problem in clinical medicine.
     The term“tissue engineering”was officially put forward in 1987 . and has been developed very quickly in recent years. Tissue engineering merges the fields of cell biology, engineering, materials science and surgery to fabricate new functional tissue using living cells and a matrix or scaffold . It is concerned with the creation of biological substitutes designed to maintain, restore or improve the function of damaged tissues and organs.
     Mesenchymal stem cells (MSCs) have been isolated not only from bone marrow, but also from many other tissues such as adipose tissue, skeletal muscle, liver, brain and pancreas. Because MSC were found to have the ability to differentiate into cells of multiple organs and systems such as bone, fat, cartilage, muscle, neurons, hepatocytes and insulin-producing cells, MSCs have generated a great deal of interest for their potential use in regenerative medicine and tissue engineering. In 2001, Zuk et al. showed that a population of adipose derived cells similar to the SVF fraction contained cells with multilineage potential. In their work, cells isolated from human lipoaspirates,termed PLA cells,underwent adipogenesis, osteogenesis, chondrogenesis, and myogenesis in vitro, suggesting that the SVF fraction of adipose may be composed not just of lineage-limited preadipocytes but of multipotent stem cells. Obtained through cosmetic procedures, such as lipoaspirates, adipose-derived mesenchymal stem cells (ADMSCs) can be harvested in large quantities, with minimal pain and morbidity incurred by the patient. This simple fact may make substantial quantities of adult stem cells available for either clinical or tissue engineering applications or for molecular characterization.
     Three-dimensional scaffold structure, which is needed to be degradable and to maintain stable volume and shape. a good scaffold must facilitate cell attachment without provoking an immune response, permit the diffusion of nutrients from the blood, and, at least initially, mimic the mechanical properties of the tissue. Additionally, the scaffold can be constructed with or designed to release factors that can beneficially manipulate the behavior of the cells in the device. Alginate, or alginic acid, is an unbranched binary copolymer of (1-->4)-linked beta-D-mannuronic acid and alpha-L-guluronic acid. Alginate readily forms binding interactions with a variety of divalent metal ions, such as Ca2+ and Ba2+. This binding has been used to cross-link bulk alginates for a wide variety of applications, particularly in areas of tissue engineering, medical devices, and wound-healing dressings.
     Regeneration of damaged cartilage in different pathological situations is a major goal which could be achieved through physiologic regeneration. The results of this study suggested that MSCs seem to be the best candidates for cell therapy for regeneration of injured tissue. They are easily isolated and can be rapidly amplified. When these cells are implanted into cartilage defects, they appear to be able to proliferate and differentiate into chondrocytes. This procedure is of considerable relevance for the regeneration of large areas of cartilage lesions. However, complete healing is difficult to obtain and requires integration of newly regenerated tissue with the surrounding host tissue and true differentiation through pathways involved in embryonic development. This goal may be achieved through the combination of MSC mediated therapy and gene transfer of selective differentiating cytokines. Objective:
     To investigate the biological features of human adipose-derived mesenchymal stem cells (ADMSCs ) to isolate and culture, then differentiate into adipocyte osteoblast and chondrocyte in vitro, and discuss the feasibility of ADMSCs as seed cells in cartilage tissue engineering. To study the feasibility of applying ADMSCs as seed cells and alginate as the scafold in constructing tissue engineering cartilage in vitro. To study the relation of seed cells, scaffold structure and microenvironment; investigate the results of the articular cartilage repaired by implantation ADMSCs suspended in calcium alginate. Methods:
     1. Human adipose was obtained from the healthy womanhood during the abdominal liposuction procedure and digested with collagenaseⅠto isolate ADMSCs , which were incubated in high-glucose DMEM with 0.10 volume fraction fetal bovine serum (FBS) for primary culture. Growth curve of the third-pas sage ADMSCs was drawn by cytometry; the express ions of CD13 and CD34 were assayed by flow cytometry and immunofluorescence technique. The third-passage ADMSCs were cultured in high-glucose DMEM supplemented with selective differentiating cytokines for adipogenic and osteogenesis induction. The appearance and generation of ADMSCs were observed everyday under inverted microscope, and the differentiations to adipocyte and osteoblast were verified by specific staining.
     2. Human adipose was obtained from the healthy womanhood during the abdominal liposuction procedure and digested with collagenaseⅠto isolate ADMSCs , which were incubated in high-glucose DMEM with 0.10 volume fraction fetal bovine serum (FBS) for primary culture. The third-passage ADMSCs were cultured in high-glucose DMEM supplemented with selective differentiating cytokines for chondrogenic induction. The appearance and generation of ADMSCs were observed everyday under inverted microscope, and the differentiations to chondrocyte were verified by AB-PSA staining and immunocytochemical staining.
     3. Applying ADMSCs as seed cells and alginate as the scafold in constructing tissue engineering cartilage in vitro. Tissue engineering cartilage were impianted subcutaneously and articular in athymic mice after cultured for 1 week in vitro. New cartilage formation was assessed though gross observation and histological examination.
     Results:
     1. ADMSCs are easily isolated and can be rapidly amplified. ADMSCs cultured in vitro were in flat fusiform shape and stably passaged, during which ADMSCs kept the same shape. The expression of stromal cell-associated marker CD13 and stem cell-associated marker CD34 was positive. The characteristics of adipocyte and osteoblast were shown after the ADMSCs had exposed to the defined medium.
     2. Chondrogenic induction of PLA cells, under micromass conditions, resulted in cell condensation after induction and was followed by ridge and spheroid/nodule formation. ADMSCs cultured in chondrogenic differentiation medium, AB-PSA staining was positive and the typeⅡcollagen immunocytochemical staining appeared positive with brown cytoplasm.The cell morphology of induced ADMSCs was evident that they obtained the differentiation phenotype as chondrogenic lineages. The expression of the typeⅡcollagen and Sox9 is an early and practically unique marker of chondrogenic differentiation. RT-PCR analysis of mRNA encoding typeⅡcollagen and Sox9 showed that high lever expression for 2 weeks.The chondrogenesis of ADMSCs were supported by presence of some other important components of cartilaginous extracellular matrix, such as typeⅨ,Ⅹ,Ⅺcollagen, AGN、COMP and Link Protein. In contrast to this, there was a trend to decrease in expression of the typeⅠcollagen after induction. Shortly after ADMSCs were seeded onto scaffolds, the calcium alginate microsphere presented the translucent globes and smooth surface. ADMSCs presented single cell condition and evenlyspred around in the microsphere. 3. In the impianted subcutaneously groups, the uninduced ADMSCs underwent fibroblast differentiation. In contrast to this, the induced ADMSCs still shown characteristic of chondrogenic. The groups with a single full thickness osteochondral injury underwent a fibrocartilaginous repair that go with serve osteoarthritis. The healing with alginate groups shown minor osteoarthritis. But the damage of articular cartilage did not heal. The healing with ADMSCs groups can significant attempt to recreate the normal articular cartilage.
     Conclusions:
     1. This study indicates that liposuction aspirates can provide abundant ADMSCs with the potential of multilineage differentiation and proliferation over extended periods. Adipose tissue can be a promising adult stem cell source. ADMSCs can be used as seed cells for cartilage tissue engineering.
     2. ADMSCs can be induced to differentiate towards chondrocytes in conditioned medium in vitro. Calcium alginate represent good compatibility and adhesion with ADMSCs without little toxicity. It appears that ADMSCs and calcium alginate microsphere may provide an excellent support for cell transplantation.
     3. The induced ADMSCs still shown characteristic of chondrogenic after impianted subcutaneously. The induced ADMSCs still shown characteristic of chondrogenic after impianted in damaged articular cartilage, and significant attempt to recreate the normal articular cartilage.
引文
1. Mankin HJ. The response of articular cartilage to mechanical injury. J Bone Joint Surg Am.1982;64:460-6.
    2. Baumbach K, Petersen JP, Ueblacker P, et al . The fate of osteochondral grafts after autologous osteochondral transplantation: a one-year follow-up study in a minipig model. Arch Orthop Trauma Surg. 2007;[Epub ahead of print].
    3. Wang Y, Blasioli DJ, Kim HJ, et al . Cartilage tissue engineering with silk scaffolds and human articular chondrocytes. Biomaterials. 2006; 27 (25) : 4434- 42.
    4. Kaneshiro N, Sato M, Ishihara M,et al . Bioengineered chondrocyte sheets may be potentially useful for the treatment of partial thickness defects of articular cartilage. Biochem Biophys Res Commun.2006;349(2):723-31.
    5. Henson FM, Vincent T. Chondrocyte outgrowth into a gelatin scaffold in a single impact load model of damage/repair - effect of BMP-2. BMC Musculoskelet Disord.2007;8(1):120.
    6. Cucchiarini M, Madry H. Gene therapy for cartilage defects. J Gene Med.2005;7(12):1495-509.
    7. Eyre DR, Weis MA, Wu JJ. Articular cartilage collagen: an irreplaceable framework? Eur Cell Mater. 2006;12:57-63.
    8. Kiani C, Chen L, Wu VJ, et al. Structure andfunction of aggrecan. Cell Res.2002;12: 19-32.
    9. 娄思权. 骨关节炎的病理与发病因素. 中华骨科杂志.1996;16(1):56.
    10. Beris AE, Lykissas MG, Papageorgiou CD, et al. Advances in articular cartilage repair. Injury. 2005;36 Suppl 4:S14-23.
    11. 王丽.同种异体移植物修复骨软骨缺损的研究现状.中国冶金工业医学杂志.2004;21(1):25-26.
    12. Hulmes DJ , Marsden ME, Strachan RK, et al. Intra-articular hyaluronate in experimental rabbit os teoarthritis can prevent changes in cartilage proteoglycan content. Osteoarthritis Cartilage.2004;12(3):232-238.
    13. Fuchs S, Monikes R, Wohlmeiner A, et al. Intra-articular hyaluronic acid compared with corticoid injections for the treatment of rhizarthrosis. Osteoarthritis Cartilage. 2006;14(1):82-88.
    14. 刘勇.转化生长因子-β与骨、软骨代谢及修复.国外医学:创伤与外科基本问题分册.1999;20(2):67270.
    15. Olah,E.H.,Kostenszky,et al.Effect of prednisolone on the glycosamino glycan components of the regenerating articular cartilage.Acta boil.1976; 27:129-134.
    16. Frank E, Grodzinsky A, Koob T,et al. Streaming potentials: A sensitive index of enzymatic degradation in articular cartilage. J Orthop Res.1987; 54:497-508.
    17. Lee RC,Frank EH,Grodzinsky AJ,et al.Oscillatory compressional behavior of articular cartilage and its associated electromechanical properties. J Biomech Eng .1981; 103:280-292.
    18. MacGinitie LA, Gluzband YA, Grodzinsky AJ. Electric field stimulation can increase protein synthesis in articular cartilage explants. J Orthop Res.1994; 12:151-160.
    19. Wang W, Wang Z, Zhang G,et al. Up-regulation of chondrocyte matrix genes and products by electric fields. Clin Orthop.2004;S163-S173.
    20. Bobacz K, Graninger WB, Amoyo L,et al. Effect of pulsed electromagnetic fields on proteoglycan biosynthesis of articular cartilage is age dependent. Ann Rheum Dis. 2006;65(7):949-51.
    21. Ciombor DM, Lester G, Aaron RK,et al. Low frequency EMF regulates chondrocyte differentiation and expression of matrix proteins. J Orthop Res.2002; 20:40-50.
    22. Brighton CT, Jensen L, Pollack SR,et al. Proliferative and syntheticresponse of bovine growth plate chondrocytes to various capacitively coupled electrical fields.J Orthop Res. 1989; 7:759-765.
    23. Salter RB ,Ogilvie-Harris DJ . The healing of intra-articular fractures with continuous passive motion. In :Louis ST ,Mosby CV. Instructional Course Lectures. American Academy of Orthopaedic Surgeons .1979 ;28 :102-117.
    24. Salter RB. The biologic concept of continuous passive motion of synovial joints. The first 18 years of basic research and its clinical application. Clin Orthop.1989;242 :12-25.
    25. Vachon A ,Branmlage LR , Gabel AA ,et al. Evaluation of the repair process of cartilage defects of the equine third carpak bone with and without subchondral bone perforation. Am J Vet Res.1986;47 :2637-2645.
    26. Kim HKW,Moran ME ,Salter RB. The potential for regeneration of articular cartilage indefects created by chondral shaving and dub-chondral abrasion. An experimental investigation in rabbits. J BoneJoint Surg.1991; 732A :1301-1315.
    27. Steadman JR, Karas SG, Miller BS,et al. Microfracture technique in the treatment of full-thickness chondral lesions of the knee in National Football League players. Am J Knee Surg. 2003;16(2):83-86.
    28. O'Driscoll SW,Keeley FW,etal.The chondrogenic potential of free autogenous periosteal grafts for biological resurfacing of major full-thickness defects in joint surfaces under the influence of continuous passive motion, An experimental investigation in the rabbit.J Bone Joint Sury,1986;68A: 1017-1035.
    29. Caplan AI etal .Principles of cartilage repair and regeneration. Clin Prthop.1997;342:254-269.
    30. Peterson L, Minas T, Brittberg M, et al. Treatment of osteochondritis dissecans of the knee with autologous chondrocyte transplantation: results at two to ten years.J Bone Joint Surg Am.2003;85-A Suppl 2:17- 24.
    31. Peterson L, Minas T, Brittberg M, et al. Two- to 9-year outcome afterautologous chondrocyte transplantation of the knee. Clin Orthop.2000; (374):212-34.
    32. Brittberg M, Lindahl A, Nilsson A, et al. Treatment of deep cartilage defects in the knee with autologous chondrocyte transplantation. N Engl J Med.1994; 331(14): 889- 95.
    33. Russlies M, Behrens P, Ehlers EM, et al. Periosteum stimulates subchondral bone densification in autologous chondrocyte transplantation in a sheep model.Cell Tissue Res.2005; 319(1): 133-42.
    34. Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells. Science.1999;284(5411): 143-7.
    35. Peterson L, Minas T, Brittberg M, et al. Two- to 9-year outcome after autologous chondrocyte transplantation of the knee. Clin Orthop.2000; (374): 212-34.
    36. Brittberg M, Peterson L, Sjogren-Jansson E, et al. Articular cartilage engineering with autologous chondrocyte transplantation. A review of recent developments.J Bone Joint Surg Am.2003;85- A Suppl 3:109-15.
    37. O’Driscoll SW, Saris DB, Ito Y, et al. The chondrogenic potential of periosteum decreases with age. J Orthop Res.2001; 19(1):95-103.
    38. Sohn DH, Lottman LM, Lum LY, et al. Effect of gravity on localization of chondrocytes implanted in cartilage defects. Clin Orthop Relat Res.2002;(394):254-62.
    39. Outerbridge HK, Outerbridge AR, Outerbridge RE.The use of a lateral patellar autologous graft for repair of a large osteochondral defect in the knee.J Bone Joint Surg(Am).1995;77:65-72.
    40. Matsusue Y,Yamamuro T,Hama H.Arthroscopic multiple osteochondral transplantation to the chondral defect in the knee associated with anterior cruciate ligament disruption.Arthroscopy.1993;9:318-321.
    41. Maranas CD,Floudas CA,Pardalos PM.New results in the packing of equal circles in a square.Discrete Mathematics.1995;142:287-293.
    42. Hangody L,Fules P.Autologous osteochondral mosaicplasty for the treatment of full-thickness defects of weight-bearing joints:ten years of experimental and clinical experience.J Bone Joint Surg Am.2003;85(Suppl 2):25-32.
    43. Bouwmeester SJ ,Beckers JM ,Kuijer R,et al. Long-term results of rib perichondrial graft for repair of cartilage defects in the human knee.Int Orthop.1997;21(5:313-317.
    44. Guo T, Zhao JN, Zhang JF, et al. Porous chitosan2gelatin scaf-fold containing plasmid DNA encoding Transforming growth factor-β1 for chondrocytes proliferation. Biomaterials. 2006;27 (7) : 1095-1103.
    45. Madry H, CucchiariniM, Terwilliger EF, et a l. Recombinant adeno-associated virus vectors efficiently and persistently transduce chondrocytes in normal and osteoarthritic human articular cartilage . Hum Gene Ther.2003 ;14 ( 4 ) :393-402.
    46. Linda J, Dwight A. Transcrip tion of bone and cartilage genes. CurrOp in Orthop.2002;13 (5) : 375-381.Ihle JN. STATs: STATs: signal transducers and activators of transcription. Cell, 1996, 84(3): 331-334
    47. Paul G, Li J Y, Brundin P. Stem cells : hype or hope ?Drug Discov Today.2002 ;7 : 295-302.
    48. Yannaki E. The papayannopoulou. Stem cells have an identity crisis. Haema. 2001 ;4 : 158-166.
    49. Friedenstein AJ , Piatetzky Shapiro II , Petrakova KV. Osteogenesis in transplants of bone marrow cells. J Embryol Exp Morphol .1966;16 : 381-390.
    50. Morasso MI,Tomic-Canic M.Epidermal stem cells:the cradle of epidermal determination, differentiation and wound healing.Biol Cell. 2005; 97: 173- 183.
    51. Berninger B, Hack MA, Gotz M. Neural stem cells: on where they hide, in which disguise, and how we may lure them out. Handb Exp Pharmacol.2006;(174):319-60.
    52. Trounson A. A fluid means of stem cell generation.Nat Biotechnol. 2007 ;25(1):62-3.
    53. Zuk PA, Zhu M, Mizuno H, et al. Multilineage cells from human adipose tissue: Implications for cell-based therapies .Tissue Eng .2001;7(2): 211-228.
    54. Rodriquez AM,Elabd C,Amri EZ, et al. The human adipose tissue is a source of multipotent stem cells. Biochimie. 2005 ;87(1):125-8.
    55. Strem BM, Hicok KC, Zhu M, et al. Multipotential differentiation of adipose tis sue-derived stem cells . Keio J Med. 2005;54(3):132-141.
    56. Xu Y, Malladi P, Wagner DR, et al. Adipose-derived mesenchymal cells as a potential cell source for skeletal regeneration. Curr Opin MolTher.2005;7(4):300-305.
    57. Yang LY, Liu XM, Sun B, et al. Adipose tis sue-derived s tromal cells express neuronal phenotypes . Chin Med J (Engl). 2004;117(3):425-429.
    58. Ogawa R, Mizuno H, Watanabe A, et al. Os teogenic and chondrogenic differentiation by adipose-derived s tem cells harves ted from GFP transgenic mice. Biochem Biophys Res Commun. 2004;313(4):871-817.
    59. Ning H, Lin G, Lue TF, et al. Neuron-like differentiation of adipose tissue- derived stromal cells and vascular smooth muscle cells . Differentiation. 2006; 74(9-10): 510-518.
    60. Watt FM , Hogan BL. Out of Eden : Stem cells and Their Niches. Science. 2000; 287: 1427-1430.
    61. Oksvold MP , Skarpen E , Widerberg J . Fluorescent histochemical techniques for analysis of intracellularsignaling. J Histochem Cytochem . 2002 ; 50 : 289-303.
    62. Herzenberg LA , De Rosa SC. Monoclonal antibodies and the FACS: complementary tools for immunobiology and medicine.Immunol Today . 2000 ; 21 : 383-390.
    63. Hui H , Perfetti R. Pancreas duodenum homeobox-1 regulates pancreas development during embryogenesis and islet cell function in adulthood . Eur J Endocrinol. 2002 ; 146 : 129-141.
    64. De Ugarte DA, Alfonso Z, Zuk PA, et al. Differential expres s ion of stem cell mobilization-as sociated molecules on multi-lineage cells from adipose tis sue and bone marrow. Immunol Lett .2003;89 (2-3):267-270.
    65. CONGET P A , MINGGUELL J J . Phenotypical and functional propertyes of human bone marrow mesenchymal progenitor cells[J ] . J Cell Physiol .1999 ;181(1) : 67 - 73.
    66. PITTINGER M F , MACKAYA M, BECK S C , et al. Multilineage potential of adult human mesenchymal stem cells. Science . 1999 ; 284(5411) : 143 - 147.
    67. BRUDER S P , RICALTON N S , BOYNTON R E , et al. Mesenchymal stem cell - surface antigen SB - 10 corresponds to activated leukocyte cell adhesion molecule and is involved in osteogenic differentiation[J ] . J Bine Miner Res . 1998 ; 13(3) : 655 - 663.
    68. De Uqarte DA,Morizono K,Elbarbarv A, et al. Comparison of multi- lineage cells from human adipose tis sue and bone marrow. Cells Tis sues Organs. 2003; 174(3): 101-109.
    69. Liu TM, Martina M, Hutmacher DW, et al. Identification of common pathways mediating differentiation of bone marrow and adipose tissue-derived human mesenchymal s tem cells into three mesenchymal lineages. Stem Cells. 2007; 25(3): 750-760.
    70. Stem cells from adipose tissue, and differentiated cells from said cells. US2004229351
    71. Mitchell JB, McIntosh K, Zvonic S, et al. Immunophenotype of human adipose-derived cells : temporal changes in s tromal-as sociated and stem cell-as sociated markers . Stem Cells .2006; 24(2): 376-385.
    72. De DA, Alfonso Z, Zuk PA, et al. Differential expression of stem cellmobilization-associated molecules on multi-lineage cells from adipose tissue and bone marrow. Immunol Lett. 2003 ;89(2-3):267-70.
    73. Gronthos S, Franklin DM, Leddy HA, et al. Surface protein characterization of human Adipose tissue-derived stromal cells. J Cellular Physiology. 2001; 189 (1): 54-63
    74. Gimble JM. Adipose tissue-derived therapeutics. Expert Opin Biol Ther. 2003 ;3(5):705-13.
    75. Zuk PA, Zhu M, Ashjian P, et al.Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002; 13(12): 4279-95.
    76. AustL, Devlin B, Foster SJ,et al. Yield of human Adipose-derived adult stem cells from liposuction aspirates. CYTOT Herapy. 2004 ; 6 (1) : 7-14.
    77. Nishimoto S,Takagi M,Wakitani S,et al.J Biosci Bioeng. 2005; 100(1): 123-126.
    78. Suuronen R, Laine P, Pohjonen T,et al. J Oral Maxillofac Surg. 1994;52(7):715-20; discussion 720-1.
    79. Stone KR,Steadman JR,Rodkey W G,et a1.Regeneration of meniscal cartilage with use of a collagen scaffold.Analysis of preliminary data.J Bone Joint Surg.1997;79(12):1770-1777.
    80. Nettles DL,Elder SH,Gilbert JA.Potentia1 use of chitosan as a cell scaffold material for cartilage tissue engineering . Tissue Engineering.2002;8(6):1009-1016.
    81. Aigner J,Tegeler J,Hutzler et a1.Cartilage tissue engineering with novel nonwoven structured biomaterial based on hyaluronic acid benzyl ester.J Biomed Master Res.1998;42 (2):172-181.
    82. 傅 捷,祝云利,吴海山,等. 兔关节软骨细胞在纤维蛋白胶中体外培养. 第二军医大学学报.2000;21 (7): 670-672.
    83. Wambach BA, Cheung H, Josephson GD. Cartilage tissue engineering using thyroid chondrocytes on a type I collagen matrix. Laryngoscope.2000;110 (12) : 2008-2011.
    84. Livingston T, Ducheyne P, Garino J In vivo evaluation of a bioactive scaffold for bone tissue engineering.J Biomed M ater Res.2002;62(1):l-l3.
    85. Ren GH, Pei GX, Gu LQ, et a1.Biocompatibility of adult human osteoblasts with coral—derived hydroxyapatite in vitro.Di Yi Junyi DaxueXuebao.2002;22(11):974-978.
    86. Langer R, Vacanti JR .Tissue engineering Science.1993; 260(5l10): 920-926.
    87. Liu Y, Chen F, LiuW, et al. Repairing large porcine full-thickness defects of articular cartilage using autologous chondrocyteengineered cartilage. Tissue Eng. 2002; 8 (4):709-721.
    88. Puelacher W C,Wisser J,Vacanti CA,et a1.Temporomandibalar joint disc replacement made by tissue—engineered growth of cartilage.J Oral Maxillofac Surg.1994;52 (11):1172-1177.
    89. Lohmann CH, Schwartz Z, Niederauer GG, et al. Pretreatment with platelet derived growth factor-β modulates the ability of costochondral resting zone chondrocytes incorporated into PLA /PGA scaffolds to form new cartilage in vivo . Biomaterials.2000;21 (1): 49-61.
    90. 杨柳. 加强软骨与骨组织工程中关键技术的应用. 中华实验外科杂志. 2005; 22 (3) : 263-265.
    91. Gerard C, Catuogno C, Amargier-Huin C, et a1. The effect of alginate,hyaluronate and hyaluronate derivatives biomaterials on synthesis of non-articccular chondrocyte extracellular matrix. JMater SciMaterMed. 2005;l6 (6) :54l-551.
    92. Liao SS, Cui FZ, Zhang W, et a1. Hierarchically biomimetic bone scaffold materials: nano-HA/collagen/PLA composite. J Biomed Mater Res BAppl Biomater. 2004 ;69(2):l58-l65.
    93. Tian WM, Hou SP, Ma J, et a1. Hyaluronic acid-poly-D-lysinebased three-dimensional hydrogel for traumatic brain injury. Tissue Eng.2005;l1(3-4):5l3-525.
    94. Grasdalen H, Larsen B, Smidsrod O. A P.M. R study of the composition and sequence of uronate residues in alginate . Carbohydr Res. 1979;68:23-31.
    95. Rees. Polysaccharide shapes and their interactions some recent advances. Pure Appl. Chem; 1981, 53:1-14.
    96. Smidsrod O, Skjak-Braek G. Alginate as immobilization matrix for cells. Trands Bioteelmol. 1990; 8 (3) :71-78.
    97. Bouhadir KH, Lee KY, Alsberg E, et al. Degradation of partially oxidized alginate and its potential application for tissue engineering. Biotechnol Prog. 2001; 17(5):945-950.
    98. Shoichet MS, Li RH, White ML, et al. Stability of hydrogels used in cell encapsulation : An in vitro comparison of alginate and agarose. Biotechnol. Bioeng , 1996;50 (4):374-381.
    99. Wee S, Gombotz WR. Protein release from aiginate matrices . Adv Drug Deliv Rev. 1998;31(3):267-285.
    100. LeRoux MA, Guilak F, Setton LA. Compressive and shear properties of alginate gel : effects of sodium ions and alginate concentration. J Biomed Mater Res. 1999;47(1) :46-53.
    101. De Vos P, De Haan BJ, Wolters GH, et al. Improved biocompatibility but Iimited graft survival after purification of alginate for microen capsulation of pancreatic islets . Diabetologia. 1997;40(3):262-270.
    102. B Kulseng, G Skj?k-Br?k, L Ryan, et al. Transplantation. 1999;67: 978-984.
    103. MarijinissenWJCM, Van Osch GJVM, Aigner J. Alginate as a chondrocyte-elivery substance in combination with a non-oven affold for cartilage tissue engineering. Biomaterials. 2002;23:1511-1517.
    104. Wang L, Shelton R M, Cooper P R, et al. Evaluation of sdium alginate for bone marrow cell tissue engineering.Biomaterials. 2003; 24(20):3475-3481.
    105. Barralet J E, Wang L, Lawson M, et al. Comparison of bone marrow cell growth on 2D and 3D alginate hydrogels.J Mater SciMaterMed. 2005;16(6):515-519.
    106. Sakai S, Masuhara H, Yamada Y, et al. Transition of mechanical property of porous alginate scaffold with cells during culture period. J Biosci Bioeng. 2005 ;100(1):127-9.
    107. Tanaka H, Mat sumara M, Veliky I A. Diffusion characteristics of subst rates in calcium alginate gel beads. Biotechol Bioeng. 1984:26 (1):53-58.
    108. Chai Yi, Mei Lehe, Lin Dongqiang, et al. Diffusion coefficient s in int rahollow calcium alginate microcapsules. J Chem Eng Data. 2004;49:475-478.
    109. Pu H T, Yang R Y K. Diffusion of sucrose and yohinobine in calcium alginate gel beads with or without ent rapped plant cells. Biotech Bioeng. 1988;32 (1):891-896.
    110. Liu L, Chen RL, Tian WD,et al. A study on the chondrogenesis of the compound of alginate gelatin and bone marrow stromal cells in vivo. Hua Xi Kou Qiang Yi Xue Za Zhi. 2005 ;23(1):60-2.
    111. Awad HA, Wickham MQ, Leddy HA,et al. Chondrogenic differentiation of adipose-derived adult stem cells in agarose, alginate, and gelatin scaffolds. Biomaterials. 2004 ;25(16):3211-22.
    112. Stevens MM, Qanadilo HF, Langer R,et al. A rapid-curing alginate gel system: utility in periosteum-derived cartilage tissue engineering. Biomaterials. 2004 ;25(5):887-94.
    113. Gründer T, Gaissmaier C, Fritz J,et al. Bone morphogenetic protein (BMP)-2 enhances the expression of type II collagen and aggrecan in chondrocytes embedded in alginate beads. Osteoarthritis Cartilage. 2004 ;12(7):559-67.
    114. Li Z, Zhang M. Chitosan-alginate as scaffolding material for cartilage tissue engineering. J Biomed Mater Res A. 2005 ;75(2):485-93.
    115. Kuo SM, Wang YJ, Weng CL,et al. Influence of alginate on type II collagen fibrillogenesis. J Mater Sci Mater Med. 2005 ;16(6):525-31.
    116. Hicks DL, Sage AB, Shelton E,et al. Effect of bone morphogenetic proteins 2 and 7 on septal chondrocytes in alginate. Otolaryngol Head Neck Surg. 2007;136(3):373-9.
    117. Alexander TH, Sage AB, Schumacher BL,et al. Human serum for tissue engineering of human nasal septal cartilage. Otolaryngol Head Neck Surg. 2006 ;135(3):397-403.
    118. Lin Y, Luo E, Chen X, Liu L,et al. Molecular and cellular characterization during chondrogenic differentiation of adipose tissue-derived stromal cells in vitro and cartilage formation in vivo. J Cell Mol Med. 2005;9(4):929-39.
    119. Leddy HA, Awad HA, Guilak F. Molecular diffusion in tissue- engineered cartilage constructs: effects of scaffold material, time, and culture conditions. J Biomed Mater Res B Appl Biomater. 2004 ; 70(2):397-406.
    120. Awad HA, Wickham MQ, Leddy HA,et al. Chondrogenic differentiation of adipose-derived adult stem cells in agarose, alginate, and gelatin scaffolds. Biomaterials. 2004 ;25(16):3211-22.
    121. Wang DW, Fermor B, Gimble JM,et al. Influence of oxygen on the proliferation and metabolism of adipose derived adult stem cells. J Cell Physiol. 2005 ;204(1):184-91.
    122. Robert P, Lanza.干细胞手册.北京: 科学出版社. 2006:425-447.
    123. Shor A.M.,Allen T.D.,Canfield,et al.Perictye derived from the retinal microvasculature undergo calcification in vitro.J Cell Sci. 1990; 97: 449-461.
    124. Doherty M.J.,Ashton B.A.,Walsh S.,et al.Vascular pericytes expressosteogenic potential in vitro and in vivo.J Bone Miner Res. 1998; 13: 828-838.
    125. Diefenderfer D.L.,Brighton C.T.Microvascular pericyes express aggrecan message which is regulated by BMP-2.Biochem Biophys Res.2000;269:172-178.
    126. Huss R.Isolation of primary and immortalized CD34- homatopoietic and mesenchymal stem cells from various sources.Stem Cells.2000;18:1-9.
    127. Lazaras H.M.,Haynesworth S.E.,Gerson S.L.,et al.J Hematother. 1997; 6: 447-455.
    128. Mitchell JB,McIntosh K,Zvonic S, et al. Immunophenotype of human adipose-derived cells : temporal changes in s tromal-as sociated and s tem cell-as sociated markers . Stem Cells .2006;24(2):376-385.
    129. Tacchetti C, Tavella S, Dozin B, et al. Cell condensation in chondrogenic differentiation. Cell Res.1992;200:26-33.
    130. Tacchetti C, Tavella S, Dozin B,et al. Cell condensation in chondrogenic differentiation. Exp Cell Res. 1992;200(1):26-33.
    131. Ueda H, Nakamura T, Yamamoto M, et al. Repairing of rabbit skull defect by dehydrothermally crosslinked collagen sponges incorporating transforming growth factor beta1. J Control Release. 2003;88(1):55-64.
    132. Steinbrech DS, Mehrara BJ, Rowe NM, et al. Gene expression of TGF-beta, TGF-beta receptor, and extracellular matrix proteins during membranous bone healing in rats. Plast Reconstr Surg. 2000 May;105(6):2028-38.
    133. Iveta D, Larry Brown, Elazer R. Cellular response to transforming growth factor beta 1 and basic fibroblast growth factor depends on release linetics and extracellular matrix interactions. J Biol Chem. 1996;271:29811-29829.
    134. Blottner D, Wolf N, Lachmund A, et al. TGF-beta rescues target-deprived preganglionic sympathetic neurons in the spinal cord.Eur J Neurosci. 1996 Jan;8(1):202-10.
    135. Chin d, Boyle GM, Parsons PG, et al. What is transforming growth factor-beta (TGF-beta)? J Plast Surg. 2004;57:215-221.
    136. Faler BJ, Macsata RA, Plummer D, et al. Transforming growth factor-beta and wound healing. Perspect Vasc Surg Endovasc Ther. 2006 Mar;18(1):55-62.
    137. N?th U, Rackwitz L, Heymer A, et al. Chondrogenic differentiation of human mesenchymal stem cells in collagen type I hydrogels. J Biomed Mater Res A. 2007 ;83(3):626-35.
    138. Mehlhorn AT, Niemeyer P, Kaiser S, et al. Differential expression pattern of extracellular matrix molecules during chondrogenesis of mesenchymal stem cells from bone marrow and adipose tissue. Tissue Eng. 2006 ;12(10):2853-62.
    139. Mehlhorn AT, Niemeyer P, Kaschte K, et al. Differential effects of BMP-2 and TGF-beta1 on chondrogenic differentiation of adipose derived stem cells. Cell Prolif. 2007 ;40(6):809-23.
    140. Sohier J, Hamann D, Koenders M, et al. Tailored release of TGF-beta1 from porous scaffolds for cartilage tissue engineering. Int J Pharm. 2007;332(1-2):80-9.
    141. Palmer GD, Steinert A, Pascher A, et al. Gene-induced chondrogenesis of primary mesenchymal stem cells in vitro. Mol Ther. 2005 ; 12(2): 219-28.
    142. Worster AA, Nixon AJ, Brower-Toland BD, et al. Effect of transforming growth factor beta1 on chondrogenic differentiation of cultured equine mesenchymal stem cells. J Vet Res. 2000 ;61(9): 1003-10.
    143. Poole CA, Flint MH, Beaumont BW. Chondrons in cartilage: Ultrastructural analysis of the pericellular microenvironment in adult human articular cartilage. J Orthop Res. 1987; 5: 509-522.
    144. Vaughan-Thomas A, Young RD, Phillips AC, et al. Characterization of type XI collagenglycosaminoglycan interactions. J Biol Chem. 2001; 276: 5303-5309.
    145. Kosher RA, Gay SW, Kamanitz JR, et al. Cartilage proteoglycan core protein genen expression during limb cartilage differentiation. Dev Biol. 1986;118:112-117.
    146. Zhou G, Zheng Q, Engin F, et al. Dominance of SOX9 function over RUNX2 during skeletogenesis. Proc Natl Acad Sci U S A. 2006 ;103(50):19004-9.
    147. Kolettas E, Muir HI, Barrett JC, et al. Chondrocyte phenotype and cell survival are regulated by culture conditions and by specific cytokines through the expression of Sox-9 transcription factor. Rheumatology (Oxford). 2001;40(10):1146-56.
    148. Bi W, Deng JM, Zhang Z, et al. Sox9 is required for cartilage formation. Nat Genet. 1999;22(1):85-9.
    149. Tsuchiya H, Kitoh H, Sugiura F, et al. Chondrogenesis enhanced by overexpression of sox9 gene in mouse bone marrow-derived mesenchymal stem cells. Biochem Biophys Res Commun. 2003 ; 301(2): 338-43.
    150. Kavalkovich KW, Boynton RE, Murphy JM,et al. Chondrogenic differentiation of human mesenchymal stem cells within an alginate layer culture system. In Vitro Cell Dev Biol Anim. 2002 ;38(8):457-66.
    151. Williams GM, Klein TJ, Sah RL. Cell density alters matrix accumulation in two distinct fractions and the mechanical integrity of alginate-chondrocyte constructs. Acta Biomater. 2005 ;1(6):625-33.
    152. Erickson GR, Gimble JM, Franklin DM, et al. Chondrogenic potential of adipose tissue-derived stromal cells in vitro and in vivo. Biochem Biophys Res Commun. 2002 ;290(2):763-9.
    153. Aydelotte MB, Thonar EJ, Mollenhauer J, et al. Culture of chondrocytesin alginate gel: variations in conditions of gelation influence the structure of the alginate gel, and the arrangement and morphology of proliferating chondrocytes. In Vitro Cell Dev Biol Anim. 1998 ;34(2):123-30.
    154. Huang JI, Zuk PA, Jones NF, Zhu M, et al. Chondrogenic potential of multipotential cells from human adipose tissue. Plast Reconstr Surg. 2004 ;113(2):585-94.
    155. Campbell JJ, Lee DA, Bader DL. Dynamic compressive strain influences chondrogenic gene expression in human mesenchymal stem cells. Biorheology. 2006;43(3-4):455-70.
    156. Benya PD, Shaffer JD, Dediefferentiated chondorcyte reexpress the differentiated collagen phenotype when cultured in agarose gels. Cell. 1982; 30:215-224.
    157. Granero-Molto F, Weis JA, Longobardi L,et al. Role of mesenchymal stem cells in regenerative medicine: application to bone and cartilage repair. Expert Opin Biol Ther. 2008 ;8(3):255-68.
    158. Salgado AJ, Oliveira JT, Pedro AJ,et al. Adult stem cells in bone and cartilage tissue engineering. Curr Stem Cell Res Ther. 2006 ;1(3): 345-64.
    159. Li WJ, Tuli R, Okafor C, Derfoul A,et al. A three-dimensional nanofibrous scaffold for cartilage tissue engineering using human mesenchymal stem cells. Biomaterials. 2005 ;26(6):599-609.
    160. Guilak F, Awad HA, Fermor B,et al. Adipose-derived adult stem cells for cartilage tissue engineering. Biorheology. 2004;41(3-4):389-99.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700