用户名: 密码: 验证码:
脂肪组织来源干细胞单克隆培养技术及其表面抗原的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
背景:
     组织工程技术以少量的种子细胞经体外扩增后与生物材料结合,修复较大的组织或器官缺损,重建缺损部位的生理功能,为临床上解决组织缺损等难题提供了一条新途径,近来受到广泛关注。
     如何获得来源广泛、充足的种子细胞是此项技术的关键。成体干细胞来源广泛、功能良好,是种子细胞的研究重点。成体干细胞可来源于骨髓、脂肪、皮肤、肌肉等各类组织,其中骨髓基质干细胞(BMSCs),发现早、取材方便、创伤小,被研究的最多。随着研究的普及和深入,脂肪组织来源干细胞(ADSCs)逐渐被人们认识,与BMSCs相比,ADSCs在体内分布更广,可利用的细胞量更多。ADSCs自1994年被发现以来已证实能向多种类型的细胞分化,并且具有取材方便、创伤小而细胞获得量大、病人易于接受的优势,在组织工程的研究和应用中显示出了巨大的潜力。
     目前普遍认为,常用方法分离出的ADSCs是由多种分化潜能不同的细胞亚群以及大量的混杂细胞组成的混合群体,其中只有部分能够形成脂肪细胞,因而必然会影响其构建脂肪组织的成功率。如何提高从脂肪组织中提取干细胞的纯度和寻找确认干细胞的相关标记等问题一直是目前急于解决的难题之一。虽然对于ADSCs的表面抗原的研究已有诸多报道,但有关ADSCs的特异性表面标志仍无统一认识,对于特定分化潜能和标志物表达之间是否存在关联至今尚未见有报道。此外,有关用于纯化细胞群获得单细胞的克隆形成试验,国外虽有利用环克隆的方法从脂肪来源细胞中分离培养出分化潜能不同的多种细胞克隆,但未对其进行相关表面抗原的分析,国内也未见有用此种方法进行干细胞研究的报道。本实验拟通过单克隆培养的方法,分离纯化出由单一细胞形成的细胞克隆,达到纯化脂肪组织来源干细胞的目的,并检测其表面抗原的表达,同时针对不同克隆鉴定其成脂分化的潜力,并检测了成脂诱导剂对ADSCs部分相关表面抗原表达的影响,寻找ADSCs成脂分化潜能和表面抗原表达之间的关联。为ADSCs在脂肪组织工程中的应用提供实验依据。
     目的:
     1.提取人体皮下脂肪组织来源干细胞(ADSCs)进行单克隆培养和扩增,以获得单克隆ADSCs,并鉴定不同克隆细胞的干细胞相关表面抗原表达。
     2.针对不同克隆ADSCs通过对其生长动力学、形态学、成脂分化能力等方面的特征进行鉴定比较,探索ADSCs成脂分化潜能和表面抗原表达之间的关联,寻找代表定向成脂分化亚群的细胞标志,从而提高构建脂肪组织的成功率。
     3.对比成脂诱导前后ADSCs部分相关表面抗原表达的变化,研究成脂诱导剂对ADSCs部分相关表面抗原表达的影响,为ADSCs在脂肪组织工程中的应用奠定实验基础。
     方法:
     临床手术切取脂肪组织,经胶原酶消化法原代培养,细胞培养并传至第2代后,有限稀释法进行克隆形成实验。针对获得的单克隆ADSCs,观察其在体外培养的形态学和生物学特点,并用流式细胞仪检测不同克隆CD29、CD34、CD44、CD54、CD106、ABCG2的表达。各克隆分别进行成脂诱导,油红O染色鉴定分化潜能。第3代的ADSCs细胞分别于成脂诱导7天前、后检测其CD34、CD54、CD106的表达。第4代ADSCs进行成脂、成骨定向诱导分化,油红“O”染色、茜素红染色定性。
     结果:
     从皮下切取的脂肪组织中能培养出大量的ADSCs,通过克隆形成实验共获得10个单克隆细胞群,不同克隆细胞在形态及增殖活力上存在差异,细胞群扩增到第9~11代后可获得细胞数2×10~6,进行后续研究及冻存。干细胞相关标志检测显示:各个克隆群体均高表达CD29(92.9±7.4%)、CD44(94.6±6.8%):低表达ABCG2(2.5±1.4%);但CD34、CD54和CD106的表达在不同群体间有明显差异。不同克隆的成脂分化潜力明显不同,油红“O”染色可见部分含红染颗粒的阳性细胞高达60%,部分为阴性结果,说明ADSCs中存在成脂分化能力不同的细胞群。第3代ADSCs在成脂诱导7天后CD34、CD54和CD106的表达较诱导之前无明显统计学差异。ADSCs成脂诱导分化两周,可见细胞内有大量脂滴,油红“O”染色可见胞浆内有大量红染颗粒;成骨诱导分化两周可肉眼看到白色矿化的颗粒状钙盐沉积,经茜素红染色鉴定可发现成骨细胞及钙盐沉积被红染。
     结论:
     本实验结果证明:皮下脂肪组织经过酶消化原代培养可提取有多向分化能力的干细胞,通过克隆形成试验培养的方法可以纯化ADSCs,获得高纯度具有间充质细胞特性的细胞群,体外多次传代、长期培养并不改变细胞形态和增殖活性。不同的单克隆细胞群之间表面标志的表达和成脂分化能力存在共性和差异。差异较显著的CD34及CD54可能作为ADSCs中高成脂分化细胞群纯化和分选的标志。另外,在成脂分化后CD34、CD54和CD106的表达较之前无显著性差异,与传代培养后的下降变化不同,也提示了这些抗原可能的标识作用。但尚不能认为这些标志物和分化能力之间存在线性关系。后续研究应当对这些标志物进行鉴定,以明确其与成脂分化之间的联系,建立脂肪组织来源基质细胞的纯化技术,为ADSCs构建脂肪组织填充物实现组织工程修复缺损奠定实验基础。
BACKGROUND:
     A soft tissue defect is generally defined as a large tissue void within the subcutaneous fat layer of the skin that may alter the contour of normal tissue. From cosmetic standard point, restoration of the soft tissue aesthetic function, rather than its physical function, is one of the primary goals that will minimize patients' anxiety and psychological stress associated with disfigurement. Millions of plastic and reconstructive surgeries are performed each year to repair soft tissue defects result from traumatic injury (i.e., significant burns), tumor resections (i.e., mastectomy and carcinoma removal), and congenital defects. Strategies to repair soft tissue defects, e.g. breast reconstruction procedures, collagen injections, and the use of autologous tissue transferplantation (i.e. free fat tissue grafts and tissue flaps), include the use of implants and fillers. However, no single filler material currently can meet these various clinical needs. Excess amounts of adipose tissue are present in many individuals with obesity, obtainable through liposuction, and can be used to transplant and repair soft tissue defects. However, resorption of the transplanted autologous tissue over time may result in 40 - 60% of the graft loss. This has significantly limited clinical application of the autologous fat tissue. Therefore, tissue-engineering strategies are being investigated and represent a novel and potential approach to generating adipose tissue.
     The primary goal of tissue engineering is to regenerate healthy tissues or organs based on patients' need, whereby eliminating tissue or organ transplantation or mechanical devices, and their related complications. Another major challenge in organ and tissue transplantation is immunological rejection after receiving allograft tissue transplantation. Evidently, tissue-engineering strategies will not be associated with these concerns. Using engineering techniques, cells collected from a health individual can be cultured and expanded to a larger cell pool. These cells can then be seeded onto a scaffold that will support cell growth and proliferation. The cell-covered on scaffold can be implanted into a site that needs surgical repair. As the cells continue growing, the scaffold material degrades or absorbs. Eventually, a new tissue mass will develop. Tissue-engineering techniques are being investigated to develop a wide range of tissues, including bone, skin, cartilage, vascular, and adipose tissues. The development of adipose tissue-engineering strategies will be essential for revolutionizing our current practice in the restoration of tissue and repair of soft tissue defects.
     Tissue engineering and regenerative medicine are an important part of the contemporary medical science that has evolved in parallel with biotechnological advances. These approaches combine biomaterials, growth factors, and stem cells to repair failing organs. Material scientists can now fabricate biocompatible scaffolds with a wide range of physical parameters, combining mechanical integrity with high porosity to promote cell infiltration and angiogenesis. Likewise, biochemists can produce highly purified, bioactive cytokines in large quantity, suitable for cell culture and in vivo applications. Despite these advances, the availability of stem cells represents the most potential approach in regenerative medicine, and technically remains very challenge before it can be used in clinical practice. By definition, a stem cell is characterized by its ability to self-renew and its ability to differentiate along multiple lineage pathways. Ideally, a stem cell for regenerative medicinal applications should meet the following criteria:
     1. Can be found in abundant quantities (millions to billions of cells)
     2. Can be harvested by a minimally invasive procedure
     3. Can be differentiated along multiple cell lineage pathways in a regulatable and reproducible manner.
     4. Can be safely and effectively transplanted to either an autologous or allogeneic host
     5. Can be manufactured in accordance with current Good Manufacturing Practice guidelines.
     There are several potential sources for obtaining stem cells for tissue regeneration or repair purposes. And the most commonly used cell types are adult and embryonic-origin. Embryonic stem cells are usually obtained from destroyed embryos, that has raised ethical concerns. On the other hand, use of stem cells derived from adult tissues provides an alternative, and avoids ethical issues related to embryonic stem cells. Mesenchymal stem cells may undergo self-renewal for several generations while remaining their specific characteristics. This type of stem cells is multipotent, easily isolated and cultured, and readily expanded in the laboratory setting. All these make mesenchymal stem cells an attractive and potential source in several clinical applications, including cell-based therapies for the diseases, such as Parkinson's and Alzheimer's diseases, spinal cord injuries, burns, heart disease, and osteoarthritis, among other conditions. These adult stem cells typically include hematopoietic stem cells, neural stem cells, bone marrow stromal cells, dermal stem cells, adipose-derived stem cells and fetal cord blood stem cells .
     Although bone marrow is the most recognized source of mesenchymal stem cells, adipose tissue has been considered as a source of multipotent cells that have the capacity of differentiating to cells of adipogenic, chondrogenic, myogenic, and osteogenic lineages when cultured with the appropriate lineage specific stimuli. Adipose-derived stem cells (ADSCs) can be obtained from tissue harvested through liposuction [termed processed lipoaspirate cells (PLAs)], or through abdominoplasty procedures. These cells have also been identified as mesenchymal cells because they are derived from adipose tissue which is, in turn, derived from mesenchyme, like bone marrow. ADSCs have been shown to be very similar to marrow-derived stem cells in morphology and phenotype. In addition to their common multipotency, several CD antigen (or cell surface) markers on the surface of marrow stem cells have also been found on the surface of ADSCs. A wide availability, easy and safe to harvest have made ADSCs a great candidate for its clinical applications in tissue-engineering. However, genetic variation of the donors and possible contamination of ADSCs by endothelial, smooth muscle, and pericyte cell populations limit the clinical application of ADSCs. Therefore, the key issue it is critically important to purify ADSCs and identify their specific surface antigens. Although several possible cell surface antigens of ADSCs have been reported, their specificity remains to be determined. Ring cloning has been used to select clones derived from a single ADSC cell. Using lineage specific differentiation media, these clones can be used to induce adipogenesis, osteogenesis, chondrogenesis, and neurogenesis. It indicated that it is possible to purify ADSCs by clone forming.
     In the present study, we established a colony unit of ADSCs, investigated the cell surface antigens of these cells, and assessed the potential of adipogenic differentiation of different ADSC clones to further determine whether adipogenesis inducement impacts the specificity of these cell surface antigens, additional assays were also performed before and after three passages of adipogenesis of the ADSCs . The present study developed a novel approach to purify the ADSCs and further characterized the cell surface antigens of these cells that will promote clinical application of ADSCs in plastic and cosmetic surgeries.
     OBJECTIVES
     1. To develop a colony unit of human adipose-derived stem cells(ADSCs), and investigate the cell-surface antigens.
     2. To determine adipogenic differentiation potential of different clones by purifying ADSCs isolated from fatty tissues, and assessing the relation of highly adipogenic potential to cell-surface antigens.
     3. To compare the changes of cell-surface antigens before and after in vitro adipogenic differentiation, find the high- adipogenic potential related cell-surface antigens
     METHODS
     ADSCs were isolated and cultivated from the resection of subcutaneous fat potions in healthy donors who under dermatoplasty. Fatty tissue were digested by collagenase, and the cells were isolated and seeded in primary culture. ADSCs were cultured for two passages , then subjected to limit dilution assays to form colony unit. Flow cytometry was used to identify the expression of cell-surface antigens in the clones obtained from above experiments. The antibodies to CD29、CD44、CD34、CD54、CD106, and ABCG2 were used to determine ADSCs specific antigens. Each clone was induced for adipogenesis, and determined by Oil Red O stain. Adipogenic, chondrogenic and osteogenic lineage differentiations of the 4th generation of ADSCs was assessed by Oil O Red, Alcian Blue, and Alizarin Red staining, respectively.
     RESULTS
     There was a large amount of ADSCs in the fatty tissue. 10 clones were obtained by colony-forming unit (CFU) assays, and after cultured for nine passages, the amount of the cells reached 2×10~6. The results of the cell-surface antigens investigation is: all of the clones were shown to be highly positive for CD29(92.9±7.4%) and CD44(94.6±6. 8%), while ABCG2 was expressed relatively low (2.5±1. 4%). The expression of CD34、CD54, and CD106 varies in different clones. Of each clone, the potential of adipogenic differentiation had a significant difference from l%~60%. Additionally, the expression of CD34, CD54, and CD106 in cells undergoing adipogenic differentiation for 7 days showed a diverse change contrast to those in cells before adipogenic differentiation. Data was analyzed by paired samples T test, and the result is no significant statistic difference. Two weeks after adipogenic differentiation of ADSCs, a significant fraction of the cells are characterized by multiple, intracellular lipid-filled droplets as indicated by Oil Red O staining, a typical function of matured fatty cells. In contrast, two weeks after chondrogenic and osteogenic induction, the cells were became positively stained by Alcian Blue and Alizarin Red, a typical function of matured chondrocyte and osteoblast cells.
     CONCLUSION
     ADSCs, harvested from adipose tissue by collagenase, is a mix population of multilineage stem cells. Clonal analysis could be used to successfully obtain purified ADSCs. Growth dynamics and morphology of the cells did not change with passages. There are differences and commonness of the expression of surface antigens among the clones. The potential of adipogenic differentiation of each clone also had variety. The difference in expression of cell surface antigens may attribute to the variation in potential of differentiation. Among those antigens identified, CD34, CD54, and CD106 showed markedly difference in each clone, maybe more associated with adipogenic differentiation clones. Additionally, the maintained expression of CD34, CD54, and CD106 after adipogenesis, while it reduced with passages, indicated that these markers may be considered as the high- adipogenic potential related cell-surface antigens. Our results suggest that the ADSCs, isolated from adipose tissue by collagenase can be the purified and certain cell surface antigens may be used to enhance ADSCs potential of adipogenicity.
引文
1.Patrick Jr CW.Tissue-engineering strategies for adipose tissue repair.Anat Rec 2001;263(4):361-6.
    2.Langstein HN,Robb GL.Reconstructive approaches in soft tissue sarcoma.Semin Surg Oncol 1999;17(1):52-65.
    3.Billings Jr E,May Jr JW.Historical review and present status of free fat graft autotransplantation in plastic and reconstructive surgery.Plast Reconstr Surg 1989;83(2):368-81.
    4. von Heimburg D, Kuberka M, Rendchen R, Hemmrich K, Rau G, Pallua N.Preadipocyte-loaded collagen scaffolds with enlarged pore size for improved soft tissue engineering. Int J Artif Organs 2003;26(12): 1064 - 76.
    
    5. Ashinoff R. Overview: soft tissue augmentation. Clin Plast Surg 2000;27(4):479-87.
    
    6. Klein AW, Elson ML. The history of substances for soft tissue augmentation.Dermatol Surg 2000;26(12):1096 - 105.
    
    7. Patrick Jr CW. Adipose tissue engineering: the future of breast and soft tissue reconstruction following tumor resection. Semin Surg Oncol 2000;19(3):302 -11.
    
    8. Katz AJ, Llull R, Hedrick MH, Futrell JW. Emerging approaches to the tissue engineering of fat. Clin Plast Surg 1999;26(4):587 - 603 viii.
    
    9. Patrick CW, Mikos AG, Mclntire LV. Frontiers in tissue engineering, 1st ed.Oxford, UK and New York: Pergamon; 1998.
    
    10. Shieh SJ,Vacanti JP.State-of the art tissue engineering:from tissue engineering to organ building[J].Surgery,2005,137(1):1-7.
    
    11. Langer R. Tissue engineering: a new field and its challenges. Pharm Res 1997;14(7):840-1.
    
    12. Fuchs JR, Nasseri BA, Vacanti JP. Tissue engineering: a 21st century solution to surgical reconstruction. Ann Thorac Surg 2001;72(2):577 - 91.
    
    13. Griffith LG. Emerging design principles in biomaterials and scaffolds for tissue engineering. Ann N Y Acad Sci 2002;961:83-95.
    
    14. Walgenbach KJ, Voigt M, Riabikhin AW, Andree C, Schaefer DJ, Galla TJ, et al.Tissue engineering in plastic reconstructive surgery. Anat Rec 2001;263(4):372 -8.
    
    15. Butler DL, Goldstein SA, Guilak F. Functional tissue engineering: the role of biomechanics. J Biomech Eng. 2000;122:570 -575.
    16. Gimble JM. Adipose tissue-derived therapeutics. Expert Opin Biol Ther. 2003;3:705-713.
    
    17. Vats A, Tolley NS, Polak JM, Buttery LD. Stem cells: sources and applications.Clin Otolaryngol Allied Sci 2002;27(4):227 - 32.
    
    18. Stem cells: a primer: National Institutes of Health. September 2002.
    
    19. Ballas CB, Zielske SP, Gerson SL. Adult bone marrow stem cells for cell and gene therapies: implications for greater use. J Cell Biochem Suppl 2002;38:20 -8.
    
    20. Conrad C, Huss R. Adult stem cell lines in regenerative medicine and reconstructive surgery. J Surg Res 2005;124(2):201 - 8.
    
    21. Habib NA. Stem cell repair and regeneration. London: Imperial College Press:Distributed by World Scientific Publishing; 2005.
    
    22. Pelled GGT, Asian H, Gazit Z, Gazit D. Mesenchymal stem cells for bone gene therapy and tissue engineering. Curr Pharm Des 2002;8(21):1917 - 28.
    
    23. Bishop AE, Buttery LD, Polak JM. Embryonic stem cells. J Pathol 2002;197(4):424 - 9.
    
    24. Snow NE. Stem cell research: new frontiers in science and ethics. Notre Dame,Ind.: University of Notre Dame Press; 2003.
    
    25. Young FE. A time for restraint. Science 2000;287(5457): 1424.
    
    26. Minguell JJ, Erices A, Conget P. Mesenchymal stem cells. Exp Biol Med (Maywood) 2001;226(6):507 - 20.
    
    27. Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, et al. Multilineage cells from human adipose tissue: implications for cellbased therapies. Tissue Eng 2001;7(2):211 -28.
    
    28. Guilak F, Lott KE, Awad HA, Cao Q, Hicok KC, Fermor B, et al. Clonal analysis of the differentiation potential of human adiposederived adult stem cells. J Cell Physiol 2006;206(1):229 - 37.
    29. Aust L, Devlin B, Foster SJ, Halvorsen YD, Hicok K, du Laney T, et al. Yield of human adipose-derived adult stem cells from liposuction aspirates. Cytotherapy 2004;6(1):7 - 14.
    
    30. Awad HA, Wickham MQ, Leddy HA, Gimble JM, Guilak F. Chondrogenic differentiation of adipose-derived adult stem cells in agarose, alginate, and gelatin scaffolds. Biomaterials 2004;25(16): 3211 - 22.
    
    31. Wang DW, Fermor B, Gimble JM, Awad HA, Guilak F. Influence of oxygen on the proliferation and metabolism of adipose derived adult stem cells. J Cell Physiol 2005;204(1):184 - 91.
    
    32. Betre H, Ong SR, Guilak F, Chilkoti A, Fermor B, Setton LA. Chondrocytic differentiation of human adipose-derived adult stem cells in elastin-like polypeptide. Biomaterials 2006;27(1): 91-9.
    
    33. Malafaya PB, Pedro AJ, Peterbauer A, Gabriel C, Redl H. Chitosan particles agglomerated scaffolds for cartilage and osteochondral tissue engineering approaches with adipose tissue derived stem cells. J Mater Sci Mater Med 2005;16:1077 - 85.
    
    34. Lin Y, Luo E, Chen X, Liu L, Qiao J, Yan Z, 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.
    
    35. Jorgensen C, Gordeladze J, Noel D. Tissue engineering through autologous mesenchymal stem cells. Curr Opin Biotechnol 2004; 15(5):406 - 10.
    
    36. Knippenberg M, Helder MN, Doulabi BZ, Semeins CM, Wuisman PI,Klein-Nulend J. Adipose tissue-derived mesenchymal stem cells acquire bone cell-like responsiveness to fluid shear stress on osteogenic stimulation. Tissue Eng2005;11(11-12):1780-8.
    
    37. Peterson B, Zhang J, Iglesias R, Kabo M, Hedrick M, Benhaim P, et al. Healing of critically sized femoral defects, using genetically modified mesenchymal stem cells from human adipose tissue. Tissue Eng 2005;11(1- 2):120 - 9.
    
    38. Strem BM, Hicok KC, Zhu M, Wulur I, Alfonso Z, Schreiber RE, et al.Multipotential differentiation of adipose tissue-derived stem cells. Keio J Med 2005;54(3):132-41.
    
    39. Tholpady SS, Llull R, Ogle RC, Rubin JP, Futrell JW, Katz AJ. Adipose tissue: stem cells and beyond. Clin Plast Surg 2006;33(1): 55 - 62 vi.
    
    40. Fraser JK, Wulur I, Alfonso Z, Hedrick MH. Fat tissue: an underappreciated source of stem cells for biotechnology. Trends Biotechnol 2006.
    
    41. Lee RH, Kim B, Choi I, Kim H, Choi HS, Suh K, et al. Characterization and expression analysis of mesenchymal stem cells from human bone marrow and adipose tissue. Cell Physiol Biochem 2004;14(4 - 6):311 - 24.
    
    42. Cheryl T. Gomillion, Karen J.L. Burg. Stem cells and adipose tissue engineering.Biomaterials 2006;27(6):52-63.
    
    43. Reinecke H, Poppa V , Murry CE. Skeletal muscle stem cells do not transd-ifferentiate into cardiomyocytes after cardiac grafting[J]. J Mol Cell Cardiol ,2002, 34(2):241-9.
    
    44. Wobus AM. Potential of embryonic stem cells[J]. Mol Aspects Med , 2001,22(3): 149-64.
    
    45. Sengenes C,Lolmede K,Zakaroff-Girard A,Preadipocytes in the human subcutaneous adipose tissue display distinct features from the adult mesenchymal and hematopoietic stem cells.J Cell Physiol. 2005 Oct;205(1):114-22.
    
    46. ThomPson JA, ltskovitz EldorJ, ShaPior SS, et al. Embryonic stem cell lines derived from human blastocytes[J]. Sciences, 1998, 282(5391): 1145-1147.
    
    47. Alhadlaq A, Tang M. Engineered adipose tissue from human mesenchymal stem cells maintains predefined shape and dimension: implications in soft tissue augmentation and reconstruction[ J ]. Tissue Eng,2005,11(3-4):556-566.
    48.Hong L,Peptan I,Clark P,et al.Ex vivo adipose tissue engineering by human marrow stromal cell seeded gelatin sponge[J].Ann Biomed Eng,2005,33(4):511-517.
    49.Mauney JR,Vladimir V,Kaplan DL.Matrix-mediated retention of adipogeni-c differentiation potential by human adult bone marrow-derived mesenchymal stem cells during ex vivo expansion.Biomaterials,2005,26(31):6167-6175.
    50.Zuk PA,Zhu M,Ashjian P,et al.Human adipose tissue is a source of multipotent stem cells[J].Mol Biol Cell,2002,13(12):4279-4295.
    51.De Ugarte DA,Morizono K,Elbarbary A et al.Comparison of multilineage cells from human adipose tissue and bone marrow[J].Cells Tissues Organs,2003,174(3):101-9
    52.Gimble JM.Adiposetissue_derivedtherapeutics[J].Expert Opin Bio Ther,2003,3(5):705_713.
    53.Sanne KB,Adrie JC,Clemens A,et al.A Rapid and Efficient Method for Expansion of Human Mesenchymal Stem Cells.Tissue Eng.2007,13(1) 3-9.
    54.朱传武.细胞问粘附分子-1与病毒性肝炎.国外医学免疫学分册,1997;20(5):262-264
    55.De Ugarte DA,Alfonso Z,Zuk PA,etc.Differential expression of stem cell mobilization-associated molecules on multi-lineage cells from adipose tissu e and bone marrow.Immunol Lett.2003 Oct 31;89(2-3):267-70.
    1.Aust L,Devlin B,Foster SJ,et al.Yield of human adipose2derived adult stem cells from liposuction aspirates.Cytotherapy,2004,6,7214.
    2.Bakker AH,Van Dielen FM,Greve JW,et al.Preadipocyte number in omental and subcutaneous adipose tissue of obese individuals.Obes Res,2004,12,4882498.
    3.Von Heimburg D,Hemmrich K,Haydarlioglu S,et al.Comparison of viable cell yield from excised versus aspirated adipose tissue.Cells Tissues Organs,2004,178,87292.
    4.鲁峰,高建华,水野博司,等.脂肪组织来源干细胞定向分化脂肪组织的体内外实验研究.中华整形外科杂志,2007,23(5):412-416
    5.Mizuno H,Zuk PA,Zhu M,Lorenz HP,Benhaim P,Hedrick MH:Myogenic differentiation by human processed lipoaspirate cells.Plast Reconstr Surg 2002;109:199-209
    6.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;290:763-769
    7.Halvorsen YD,Franklin D,Bond AL,et al.Extracellular matrix mineralization and osteoblast gene expression by human adipose tissue-derived stromal cells.Tissue Eng 2001;7:729-741
    8.Nathan S,Das DS,Thambyah A,Fen C,Goh J,Lee EH:Cellbased therapy in the repair of osteochondral defects: a novel use for adipose tissue. Tissue Eng 2003;9:733 - 744
    
    9. Planat-Benard V, Silvestre JS, Cousin B, et al. Plasticity of human adipose lineage cells toward endothelial cells: physiological and therapeutic perspectives.Circulation 2004; 109:656 - 663
    
    10. Rangappa S, Fen C, Lee EH, Bongso A, Wei ES: Transformation of adult mesenchymal stem cells isolated from the fatty tissue into cardiomyocytes. Ann Thorac Surg 2003; 75:775 - 779
    
    11. Safford KM, Safford SD, Gimble JM, et al. Characterization of neuronal/glial differentiation of murine adipose-derived adult stromal cells. Exp Neurol 2004;187:319-328
    
    12. Zuk PA,Zhu M,M izuno H,et al.Multilineage cells from human adipose tissue:implications for cell-based therapies. Tissue Eng.2001;7(2):211-228.
    
    13. Clark DL, Johansson CB, Wilbertz J, et al. Generalized potential of adult neural stem cells. Science 2000,288:1660-3.
    
    14. Guilak F, Lott KE, Awad HA, et al. Clonal analysis of the differentiation potential of human adipose-derived adult stem cells. J Cell Physiol. 2006;206(1):229-237.
    
    15. Li TS, Ito H, Hayashi M, Furutani A, et al. Cellular expression of integrin-beta(1) is of critical importance for inducing therapeutic angiogenesis by cell implantation. Cardiovasc Res 2005; 65: 64 - 72
    
    16. Brian MS, Kevin CH, Min Z, et al. Multipotential differentiation of adipose tissue-derived stem cells. Keio J Med 2005; 54 (3): 132 - 141
    
    17. Goodell MA, Brose K, Paradis G et al. Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo. J Exp Med 1996;183:1797-1806.
    
    18. Mitchell JB, Mclntosh K, Zvonic S, et al. Immunophenotype of human adipose-derived cells:temporal changes in stromal-associated and stem cell-associated markers[J].Stem Cells,2006,24(2):376-85.Epub 2005 Dec 1.
    19.Sudhoff T,Sohngen D:Circulating Endothelial Adhesion Molecules(sE-selectin,sVCAM-1 and slCAM-1) During rHuGCSF-Stimulated Stem Cell Mobilization.J Hematother Stem Cell Res 2002;11:147-151
    20.朱传武.细胞间粘附分子-1与病毒性肝炎.国外医学免疫学分册,1997;20(5):262-264
    21.De Ugarte DA,Alfonso Z,Zuk PA,etc.Differential expression of stem cell mobilization-associated molecules on multi-lineage cells from adipose tissu e and bone marrow.Immunol Lett.2003 Oct 31;89(2-3):267-70.
    22.Wang S,Yang TS,Han BS,et al.An easy method to discover cell membrane antigen with atomic force microscopy.Mol Biol Rep DOI 10.1007/s11033-007-9122-2
    1.Aust L,Devlin B,Foster S J,et al.Yield of Human Adipose-derived Adult Stem Cells from Liposuction Aspirates[J].Cytotherapy,2004,6(1):7-14.
    2.Zuk PA,Zhu M,M izuno H,et al.Multilineage cells from human adipose tissue:implications for cell-based therapies.Tissue Eng.2001;7(2):211-228.
    3.Zuk PA,Zhu M,Ashjian P,De Ugarte DA,Huang JI,Mizuno H,Alfonso ZC,Fraser JK,Benhaim P,Hedrick MH.Human adipose tissue is a source of multipotent stem cells.Mol Biol Cell.2002;13:4279-4295.
    4.De Ugarte DA,Alfonso Z,Zuk PA,et al.Differential expression of stem cell mobilization-associated molecules on multi-lineage cells from adipose tissue and bone marrow[J].Immunol Lett,2003,89(2-3):267-70.
    5.Philipp Niemeyer,Martin Komacker,Alexander Mehlhom,et al.Comparison of Immunological Properties of Bone Marrow Stromal Cells and Adipose Tissue-Derived Stem Cells Before and Aider Osteogenic Differentiation In Vitro.Tissue Engineering,2007,13(1):111-121.
    6.James B.Mitchell,Mclntosh K,Zvonic S,et al.Immunophenotype of human adipose-derived cells:temporal changes in stromal-associated and stem cell-associated markers[J].Stem Cells,2006,24(2):376-85.Epub 2005 Dec 1.
    7. Campagnoli C, Roberts IA, Kumar S et al. Identification of mesenchymal stem/progenitor cells in human first-trimester fetal blood, liver, and bone marrow.Blood 2001;98:2396 -2402.
    
    8. In' t Anker PS, Scherjon SA, Kleijburg-van der Keur C et al. Amniotic fluid as a novel source of mesenchymal stem cells for therapeutic transplantation. Blood 2003;102:1548 - 1549.
    
    9. Nakahara H, Dennis JE, Bruder SP et al. In vitro differentiation of bone and hypertrophic cartilage from periosteal-derived cells. Exp Cell Res 1991;195:492 - 503.
    
    10. Zuk PA, Zhu M, Ashjian P et al. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell 2002;13:4279 - 4295.
    
    11. Javazon EH, Beggs KJ, Flake AW. Mesenchymal stem cells: Paradoxes of passaging. Exp Hematol 2004;32:414 - 425.
    
    12. Haynesworth SE, Baber MA, Caplan AI. Cell surface antigens on human marrow-derived mesenchymal cells are detected by monoclonal antibodies. Bone 1992;13:69 - 80.
    
    13. Galmiche MC, Koteliansky VE, Briere J et al. Stromal cells from human long-term marrow cultures are mesenchymal cells that differentiate following a vascular smooth muscle differentiation pathway. Blood 1993;82:66-76.
    
    14. Pittenger MF, Mackay AM, Beck SC et al. Multilineage potential of adult human mesenchymal stem cells. Science 1999;284:143 - 147.
    
    15. Conget PA, Minguell JJ. Phenotypical and functional properties of human bone marrow mesenchymal progenitor cells. J Cell Physiol 1999;181:67-73.
    
    16. Sordi V, Malosio ML, Marchesi F et al. Bone marrow mesenchymal stem cells express a restricted set of functionally active chemokine receptors capable of promoting migration to pancreatic islets. Blood 2005;106:419 - 427.
    17. Le Blanc K, Tammik C, Rosendahl K et al. HLA expression and immunologic properties of differentiated and undifferentiated mesenchymal stem cells. Exp Hematol 2003;31:890-896.
    
    18. Baddoo M, Hill K, Wilkinson R et al. Characterization of mesenchymal stem cells isolated from murine bone marrow by negative selection. J Cell Biochem 2003;89:1235 - 1249.
    
    19. Jones EA, Kinsey SE, English A et al. Isolation and characterization of bone marrow multipotential mesenchymal progenitor cells. Arthritis Rheum 2002;46:3349 - 3360.
    
    20. Gindraux F, Selmani Z, Obert L et al. Human and rodent bone marrow mesenchymal stem cells that express primitive stem cell markers can be directly enriched by using the CD49a molecule. Cell Tissue Res 2007; 327:471 - 483.
    
    21. S. Gronthos, P.J. Simmons, S.E. Graves, P.G Robey, Integrinmediated interactions between human bone marrow stromal precursor cells and the extracellular matrix, Bone 28 (2001) 174/181.
    
    22. K. Miyake, K.L. Medina, S. Hayashi, S. Ono, T. Hamaoka, P.W. Kincade,Monoclonal antibodies to Pgp-1 /CD44 block lymphohemopoiesis in long-term bone marrow cultures, J. Exp. Med. 171 (1990) 477/488.
    
    23. X.D. Chen, H.Y. Qian, M.C. Horowitz, Cells in the osteoblast lineage express and are activated by the Thy-1 differentiation antigen, J. Bone Miner. Res. 10(1995)S308(abstract).
    
    24. F. Barry, R. Boynton, M. Murphy, J. Zaia, The SH-3 and SH-4 antibodies recognize distinct epitopes on CD73 from human mesenchymal stem cells, Biochem. Biophys. Res. Commun. 289 (2001) 519/524.
    
    25. P.J. Simmons, B. Torok-Storb, Identification of stromal cell precursors in human bone marrow by a novel monoclonal antibody, STRO-1, Blood 78 (1991) 55/62.
    26. S. Gronthos, S.E. Graves, S. Ohta, P.J. Simmons, The STRO-1_/fraction of adult human bone marrow contains the osteogenic precursors, Blood 84 (1994) 4164_/4173.
    
    27. J.E. Dennis, J.P. Carbillet, A.I. Caplan, P. Charbord, The STRO-1_/ marrow cell population is multipotential, Cells Tissues Organs 170 (2001) 73/82.
    
    28. P.A. Conget, J.J. Minguell, Phenotypical and functional properties of human bone marrow mesenchymal progenitor cells, J. Cell Physiol. 181 (1999) 67/73.
    
    29. K. Steward, S. Walsh, J. Screen, C.M. Jefferiss, J. Chainey, GR. Jordan, J.Beresford, Further characterization of cells expressing STRO-1 in cultures of adult human bone marrow stromal cells, J. Bone Mineral Res. 14 (1999)1345_/1356.
    
    30. M.K. Majumdar, M.A. Thiede, J.D. Mosea, M. Moorman, S.L. Gerson,Phenotypic and functional comparison of cultures of marrow-derived mesenchymal stem cells (MSCs) and stromal cells, J. Cell Physiol. 176 (1998) 57_/66.
    
    31. M.K. Majumdar, V. Banks, D.P. Peluso, E.A. Morris, Isolation, characterization,and chondrogenic potential of human bone marrow-derived multipotential stromal cells, J. Cell Physiol. 195 (2000) 98_/106.
    
    32. R. Kronenwett, S. Martin, R. Haas, The role of cytokines and adhesion molecules for mobilization of peripheral blood stem cells, Stem Cells 18 (2000) 320.
    
    33. P.J. Simmons, B. Masinovsky, B.M. Longenecker, R. Berenson, B. Tarok-Storb,W.M. Gallatin, Vascular cell adhesion molecule-1 expressed by bone marrow stromal cells mediates the binding of hematopoietic progenitor cells, Blood 80(1992)388_/395.
    
    34. J.B. Levesque, Y. Takamatsu, S.K. Hilsson, D.N. Haylock, R.J. Simmons,Vascular cell adhesion molecule-1 (VCAM-1) is cleaved by neutrophil proteases in the bone marrow following haematopoietic cell mobilization by granulocyte colony stimulating factor,Blood 98(2001) 1289_/12990.
    35.张明伟,于晓妉,郭子宽等,体外长期培养对人骨髓间充质干细胞生物学特性的影响,组织工程与重建外科杂志,2005,1(1):51-53。

© 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