人E-钙粘素融合蛋白基质构建干细胞微环境的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
细胞与细胞、细胞与细胞外基质之间存在着复杂的联系,细胞表面的受体与细胞外微环境相互作用,决定了细胞的粘附、增殖、迁移、分化,以及特异性功能表达。近年来,组织工程和再生医学的研究取得了重要的进展,为各领域的研究拓宽了一条新的途径。利用生物材料、医用材料等仿生构建细胞外微环境的应用已经成为生物材料领域研究的重点,其中,以基因工程技术为基础的新型生物材料---融合蛋白的研制已成为设计和构建细胞外微环境生物材料研究的新方向。本论文通过生物合成人E-钙粘素融合蛋白,研究开发仿生细胞-细胞间相互作用的生物材料,并仿生构建细胞外微环境调控细胞行为,其不仅能有效推动基础生物学的研究,同时在组织工程和再生医学等领域也有广泛的应用。
     首先,我们重组了人细胞间粘附分子E-钙粘蛋白(hE-cadherin)的胞外域与免疫球蛋白G(IgG)的Fc域,生物合成人E-钙粘素融合蛋白(hE-cadherin-Fc)。基因工程技术构建了融合蛋白表达载体,利用293F细胞表达体系进行了融合蛋白的表达并纯化,Elisa和western blotting检测表明我们成功制备了高纯度的hE-cadherin-Fc。进一步将融合蛋白应用于疏水材料的表面修饰,仿生构建了细胞-细胞间相互作用的生物材料细胞外微环境并用于人骨髓间充质干细胞(hMSCs)的增殖及定向分化研究。接触角(WCA)及原子力显微镜(AFM)检测表明:hE-cadherin-Fc能够利用Fc结构域稳定的吸附在疏水材料表面,同时改善了材料表面的形态及亲水性,形成稳定的细胞外基质。与组织细胞培养板(TC-PS)和明胶(Gelatin)基质相比,hE-cadherin-Fc基质能显著增强hMSCs的粘附,并促进细胞增殖;同时未分化性标志物CD105的流式细胞术鉴定显示:hMSCs保持了良好的未分化性。hE-cadherin-Fc基质对hMSCs生物学行为的调控及其分子机制的进一步研究,发现融合蛋白基质显著提高了E钙粘素信号通路的相关分子表达水平,初步揭示了在细胞培养初期hE-cadherin-Fc基质与细胞间的相互作用代替了细胞-细胞间的相互作用。
     其次,我们考察了hE-cadherin-Fc基质对hMSCs向肝细胞定向诱导分化的影响。在hE-cadherin-Fc基质表面hMSCs向肝细胞诱导分化培养4周后,细胞形态、肝特异性标志物基因和蛋白的表达、肝细胞分化功能表达的检测表明:不同基质条件下hMSCs均有部分细胞分化成为肝细胞样细胞,与TC-PS和Gelatin基质相比,hE-cadherin-Fc基质表面分化的肝细胞样细胞表达肝前体细胞标志物CD117的阳性细胞比例高于TC-PS和Gelatin基质表面,并且肝细胞特异性标志物基因白蛋白(ALB)、细胞角蛋白18(CK18)和肝细胞核因子4(HNF4)的分子水平表达量更高;ALB蛋白免疫荧光染色结果也证明了ALB蛋白表达与其分子表达水平相同的结果。同时,肝分化功能指标糖原储存、吲哚青绿(ICG)摄取、白蛋白和尿素分泌的检测结果表明,hE-cadherin-Fc基质显著提高了人骨髓间充质干细胞源肝样细胞分化功能的表达。本研究结果表明hE-cadherin-Fc基质与相关液性因子协同作用促进hMSCs向肝细胞定向跨胚层分化的潜能,其分子机制还有待进一步深入研究,揭示了生物材料设计对其生物学功能的影响及应用前景。
     最后,我们进一步探索了hE-cadherin-Fc用于纳米纤维支架和多孔支架的生物功能改性的研究。扫描电子显微镜(SEM)的结果显示,融合蛋白的修饰没有改变支架的三维微纳米结构。X射线光电子能谱(XPS)和Elisa检测结果表明,hE-cadherin-Fc在三维支架表面有效形成了稳定的融合蛋白层。WCA结果显示hE-cadherin-Fc表面修饰显著改善了三维支架表面的亲水性;同时,hE-cadherin-Fc基质化提高了hMSCs在三维支架上的粘附与增殖。细胞骨架染色和细胞形态结果表明,在hE-cadherin-Fc表面改性支架内的细胞更接近成纤维状。此外,茜素红染色表明hE-cadherin-Fc基质表面扩增的干细胞保持了良好的分化性能。本研究表明hE-cadherin-Fc可用于三维支架表面改性,提高支架与细胞的亲和性,进一步赋予支架生物功能,显示了其在干细胞三维环境构建及生物医学领域的应用前景。
     综上所述,hE-cadherin-Fc融合蛋白可用于疏水材料的表面改性,由钙粘素的同源结合介导细胞与材料粘附,通过生物材料模拟细胞间结合仿生构建人工细胞外微环境,显著改善干细胞粘附与增殖,并促进干细胞定向分化调控,为生物材料在再生医学领域的应用开辟了新的思路。
Extracellular matrix (ECM), neighboring cells and soluble factors are most important effectors of cell behavior and function. Among them, cell-matrix interaction plays a fundamental role in regulating cell adhesion, migration, proliferation, differentiation and the expression of specific functions. In recent years, there was some important progress in tissue engineering and regenerative medicine, which widened a new theory in various research fields. The recombinant fusion proteins, an engineered artificial ECM, which was based on gene engineering technology, has gained extensive attention because of its selectivity to specific cell lineages and tissue responses. In this study, human E-cadherin-Fc fusion protein was constructed and used as a cell-cell adhesion biomimicking matrix for human mesenchymal stem cells (hMSCs). It can not only be used in basic biological research, but also in tissue engineering and regenerative medicine.
     Firstly, a fusion protein consisting of human E-cadherin extracellular domain and the immunoglobulin G Fc region (hE-cadherin-Fc) was biosynthesized by gene engineering technology. The hE-cadherin-Fc fusion protein expressed and purified by the Free-Style MAX293Expression System was analyzed by Elisa and western blotting. Water contact angles (WCA) and Atomic Force Microscope (AFM) showed that the hE-cadherin-Fc was stably immobilized onto a polystyrene plate due to the hydrophobicity of the Fc domain, enhancing the surface wettability and topography. The hE-cadherin-Fc matrix markedly promoted the cell adhesion and proliferation of hMSCs comparing with the tissue culture-treated plate (TC-PS) and the gelatin-coated plate. Furthermore, the expanded hMSCs on the hE-cadherin-Fc were positive for CD105, showing the hE-cadherin-Fc matrix could maintain the undifferentiation of hMSCs. Additionally, the expressions of E-cadherin and β-catenin in the hMSCs were improved on the hE-cadherin-Fc matrix, suggesting that the cell-cell adhesion junctions were substituted by the interactions between the hE-cadherin-Fc matrix and the hMSCs during the initial culture stage in the absence of cell-cell interactions.
     Secondly, we focused on the effects of the hE-cadherin-Fc matrix on the hepatic differentiation from hMSCs. After4weeks of directly differentiation on modified surfaces, hepatocyte-like cells were identified through cell morphology, RT-PCR, flow cytometry, immunofluorescence, Periodic acid-Shiff staining, indocyanine green (ICG) uptake and hepatocellular synthesis and metabolism functions by albumin/urea assays. Compared with the TC-PS and gelatin-coated surface, the expression of hepatic lineage surface markers CD117was higher on hE-cadherin-Fc surface. The mRNA expressions of hepatocyte-specific markers, such as ALB, CK18and HNF4, were performed by RT-PCR. The expression levels of these markers were found to be significantly upregulated during the differentiation on hE-cadherin-Fc surface. Similarly, the expression of ALB protein which was detected by immunofluorescence staining appeared to be higher than TC-PS and gelatin-coated surface. To confirm whether differentiated hepatocyte-like cells derived from hMSC were functionally competent, we examined the glycogen storage, ICG uptake, albumin secretion and urea production. It demonstrated that the differentiated cells expressed higher function on hE-cadherin-Fc-coated surface. These results show that the hE-cadherin-Fc may be a promising artificial extracellular matrix (ECM) for the hMSCs differentiation via the homophilic interaction of hE-cadherin and the synergy between cell adhesion molecular and growth factors.
     Lastly, we fabricated nanofibrous scaffolds and porous scaffolds, modified with hE-cadherin-Fc fusion protein and studied the synergistic effect of the scaffolds. The morphology of three-dimensional scaffolds which modified by hE-cadherin-Fc fusion protein was not changed by using scanning electron microscopy (SEM). Surface modification was investigated via monitoring the changes of the surface by X-ray photoelectron spectroscopy (XPS) and Elisa showed that the hE-cadherin-Fc was stably modified on three-dimensional scaffolds. WCA measurements confirmed that the hE-cadherin-Fc fusion protein could improve the surface properties of scaffolds. The hE-cadherin-Fc matrix markedly promoted the cell adhesion and proliferation of hMSCs on three-dimensional scaffolds. The cell morphology was evaluated by F-actin staining, and the cells had a more spreading morphology on hE-cadherin-Fc-coated scaffolds. The results demonstrated that the hE-cadherin-Fc improved the cell compatibility and biological functions of three-dimensional scaffolds.
     In summary, hE-cadherin-Fc was successfully prepared as a cell-cell adhesion biomimicking matrix which can be used for the surface modification of hydrophobic materials effectively. The hE-cadherin-Fc fusion protein matrix could enhance the adhesion and proliferation of the hMSCs, and improve the differentiation regulation of stem cells. The hE-cadherin-Fc was shown to be a promising artificial ECM for hMSCs.
引文
[1]Langer R, Vacanti JP. Tissue engineering. Science,1993,260(5110):920-926.
    [2]Levenberg S, Langer R. Advances in tissue engineering. Curr Top Dev Biol,2004,61(1): 113-134.
    [3]Nasseri BA, Ogawa K, Vacanti JP. Tissue engineering:an evolving 21st-century science to provide biologic replacement for reconstruction and transplantation. Surgery,2001,130(5): 781-784.
    [4]Hench LL, Polak JM. Third-generation biomedical materials. Science,2002,295 (5557): 1014-1017.
    [5]Vacanti CA, Paige KT, Kin WS, et al. Experimental tracheal replacement using tissue-engineered cartilage. J Pediatr Surg,1994,29(2):201-204.
    [6]Hellman KB. Biomedical applications of tissue engineering technology:Regulatory tissue. Tissue engineering,1995,1(2):203-210.
    [7]Risbud M. Tissue engineering:implications in the treatment of organ and tissue defects. Biogerontology,2001,2(2):117-125.
    [8]Mikos AG, Herring SW, Ochareon P, et al. Engineering complex tissues. Tissue Engineering, 2006,12(12):3307-3339.
    [9]Atala A, Koh CJ. Tissue engineering applications of therapeutic cloning. Annu Rev Biomed Eng,2004,6(1):27-40.
    [10]Gross H. Tissue engineering--synthetic tissues proven in clinical applications. Deutsche medizinische Wochenschrift,2010,135(7):6.
    [11]Chiari C, Koller U, Lang S, et al A tissue engineering approach to meniscus regeneration in a sheep model. Osteoarthritics and Cartilage,2006,14(10):1056-1065.
    [12]Humes HD. Tissue engineering of a bioartificial kidney:a universal donor organ. Transplant Proc,1996,28(4):2032-2035.
    [13]Bettinger CJ. Synthetic biodegradable elastomers for drug delivery and tissue engineering. Pure and Applied Chemistry,2011,83(1):9-24.
    [14]Eppley BL, Dadvand B. Injectable soft tissue fillers:clinical overview. Plastic and Reconstructive Surgery,2006,118(4):98-106.
    [15]Shinoka T, Shum Tim D, Ma PX, et al. Creation of viable pulmonary artery autografts through tissue engineering. Thorac Cardiovasc Surg,1998,115(3):536-545.
    [16]Gomes S, Leonor IB, Mano JF, et al. Natural and genetically engineered proteins for tissue engineering. Progress in Polymer Science,2012,37(1):1-17.
    [17]Chen GP, Ushida T, Tateishi T. Development of biodegradable porous scaffolds for tissue engineering. Materials Science & Engineering C-Biomimetic and Supramolecular Systems, 2001,17(1-2):63-69.
    [18]Noly CE, Cheng C, Davies JE. Optimizing the sterilization of LGA scaffolds for use in tissue engineering. Biomaterials,2001,22(1):25-31.
    [19]Vacanti JP, Morse MA, Saltzman WM, et al. Selective cell transplantation using bioabsorbable artificial polymers as matrices. J Pediatr Surg,1988,23(1 Pt 2):3.
    [20]Bianco P and Robey PG. Stem cells in tissue engineering. Nature,2001,414(6859):118-121.
    [21]Taboas JM, Kuo CK, Huang GTJ, et al. Growth factors in adult stem cell-based skeletal tissue engineering, in:Mao JJ, Vunjak Novakovic G, Mikos A, et al, eds. Translational Approaches in Tissue Engineering and Regenerative Medicine,2008(1):83-124.
    [22]Pearson RG, Bhandari R, Quirk RA, et al. Recent advances n tissue engineering:an invited review. J Long Term Effed Implants,2002,12(1):1-33.
    [23]Monzack EL, Rodriguez KJ, McCoy CM, et al. Natural materials in tissue engineering applications, in:Burdick JA, Mauck RL, eds. Biomaterials for Tissue Engineering Applications:A Review of the Past and Future Trends,2011:209-241.
    [24]Peter SJ, Miller MJ, Mikos AG, et al. Polymer concepts in tissue engineering. Journal of Biomedical Materials Research (Applied Biomaterials),1998,43(4):422-427.
    [25]Dutta PK, Rinki K, Dutta J. Chitosan:A promising biomaterial for tissue engineering scaffolds, in:Jayakumar R, Prabaharan M, Muzzarelli RAA, eds. Chitosan for Biomaterials, 2011:45-79.
    [26]Chung S and King MW. Design concepts and strategies for tissue engineering scaffolds. Biotechnology and Applied Biochemistry,2011,58(6):423-438.
    [27]Sh ieh SJ, Vacanti JP. State of the art tissue engineering:from tissue engineering to organ build ing. Surgery,2005,137:1-7.
    [28]Yarlagadda PK, Chandrasekharan M, Shyan JY. Recent advances and current developments in tissue scaffolding. Mater Eng,2005,15:159-177.
    [29]Tsang VL, Bhatia SN. Three dimensional tissue fabrication. Advanced Drug De livery Reviews,2004,56(11):1635-1647.
    [30]Bongso A, Fong CY, Gauthaman K. Taking stem cells to the clinic:major challenges. J Cell Biochem,2008,105:1352-1360.
    [31]Lavik E, Langer R. Tissue engineering:current state and perspectives. Appl Microbiol Biotechnol,2004,65(1):1-8.
    [32]Griffith LG, Naughton G. Tissue engineering-current challenges and expanding opportunities. Science,2002,295(5557):1009-1014.
    [33]Wang M. Developing bioactive composite materials for tissue replacement. Biomaterials, 2003,24(13):2133-2151.
    [34]Atala A. Tissue engineering, stem cells and cloning:current concepts and changing trend s. E xpert Opin Biol Ther,2005,5(1):879-892.
    [35]Nichols JE, Niles JA, Cortiella J. Design and development of tissue engineered lung: Progress and challenges. Organogenesis,2009,5(2):57-61.
    [36]Simpson DG and Bowlin GL. Tissue-engineering scaffolds:can we re-engineer mother nature? Expert Review of Medical Devices,2006,3(1):9-15.
    [37]Dubok VA. Bioceramics yesterday, today, tomorrow. Powder Metallurgy and Metal Ceramics, 2000,39(7-8):381-394.
    [38]Cowin SC. How does nature build a tissue? in:Guilak F, Butler DL, Goldstein SA, Mooney DJ, eds. Functional Tissue Engineering,2003:3-16.
    [39]Griffith LG, Naughton G. Tissue engineering-current challenges and expanding opportunities. Science,2002,295(5557):1009-1014.
    [40]Dennis E. Discher, et al. Growth factors, matrices, and forces combine and control stem cells. Science,2009,324(26):1672-1677.
    [41]Dutta RC, Dutta AK. Cell-interactive 3D-scaffold:advances and applications. Biotechnol Adv,2009,27(4):334-339.
    [42]Rossello R A. Cell communication and tissue engineering. J Commun Integr Biol,2010,3(1): 53-56.
    [43]Rosso F, Giordano A, Barbarisi M, Barbarisi A. From cell-ECM interactions to tissue Engineering J Cell Physiol,2004,199(2):174-80.
    [44]Battista S, Guarnieri D, Netti PA, et al. The effect of matrix composition o f 3D constructs on embryonic stem cell differentiation. Biomaterials,2005,26 (31):6194-6207.
    [45]Ruckenstein E, Li ZF. Surface modification and functionalization through the self-assembled monolayer and graft polymerization. Advances in Colloid and Inter face Science,2005, 113(1):43-63.
    [46]Liu X, Won Y, Ma PX. Surface modification of interconnected porous scaffolds. Journal of Biomedical Materials Research Part A,2005,74(1):84-91.
    [47]Kim BS, Mooney DJ. Development of biocompatible synthetic extracellular matrices for tissue engineering. Trends in Biotechnology,1998,16(5):224-230.
    [48]Place ES, Evans ND, Stevens MM. Complexity in biomaterials for tissue engineering. Nat Mater,2009,8(6):457-70.
    [49]Lutolf MP, Hubbell JA. Synthetic biomaterials as instructive extracellular microenvironments for morphogenesis in tissue engineering. Nat Biotechnol,2005,23(1):47-55.
    [50]Goldberg M, Langer R, Jia X. Nanostructured materials for applications in drug delivery and tissue engineering. J Biomater Sci Polym Ed,2007,18(3):241-268.
    [51]Suh JKF, M atthew HWT. Application of chitosan based poly saccharine biomaterials in cartilage tissue engineering:a review. Biomaterials,2000,21(24):2589-2598.
    [52]Wang H, van Blitterswijk CA. The role of three-dimensional polymeric scaffold configuration on the uniformity of connective tissue formation by adipose stromal cells. Biomaterials,2010,31(15):4322-4329.
    [53]Lee KY, Mooney DJ. Hydrogels for tissue engineering. Chem Rev,2001,101(7):1869-1879.
    [54]Tampieri A, Sandri M, Quarto R, et al. Design of graded biomimetic osteochondral composite scaffolds. Biomaterials,2008,29(26):3539-3546.
    [55]Nagaoka M, Jiang H L, Hoshiba T, et al. Application of recombinant fusion proteins for tissue engineering. Annals of Biomedical Engineering,2010,38(3):683-693.
    [56]Romano NH, Sengupta D, Chung C, et al. Protein-engineered biomaterials:Nanoscale mimics of the extracellular matrix. Biochim Biophys Acta,2011,1810(3):339-49.
    [57]Arai R, Ueda H, Kitayama A, et al. Design of the linkers which effectively separate domains of a bifunctional fusion protein. Protein Eng,2001,14(8):529-532.
    [58]Straley K S, Heilshora S C. Independent tuning of multiple biomaterial properties using protein engineering. Soft Matter,2009,5(1):114-124.
    [59]Choi BH, Choi YS, Kang DG, et al. Cell behavior on extracellular matrix mimic materials based on mussel adhesive protein fused with functional peptides. Biomaterials,2010,31(34): 8980-8988.
    [60]Nagaoka M, Koshimizu U, Yuasa S, et al. E-cadherin-coated plates maintain pluripotent es cells without colony formation. Plos One,2006,1(1):e15.
    [61]Amranul Haque, Xiao-Shan Yue, Toshihiro Akaike, et al. Characterization and neural differentiation of mouse embryonic and induced pluripotent stem cells on cadherin-based substrata. Biomaterials,2012,33(20):5094-5106.
    [62]Yue X-S, Murakami Y, Tamai T, et al. A fusion protein N-cadherin-Fc as an artificial extracellular matrix surface for maintenance of stem cell features. Biomaterials,2010,31(20): 5287-5296.
    [63]Qingyuan Meng, Amranul Haque, Toshihiro Akaike, et al. The differentiation and isolation of mouse embryonic stem cells toward hepatocytes using galactose-carrying substrata. Biomaterials,2012,33(5):1414-1427.
    [64]Ogiwara K, Nagaoka M, Cho C-S, et al. Construction of a novel extracellular matrix using a new genetically engineered epidermal growth factor fused to IgG-Fc. Biotechnology Letters, 2005,27(20):1633-1637.
    [65]Masato Nagaoka, Yuko Hagiwara, Toshihiro Akaike, et al. Design of the artificial acellular feeder layer for the efficient propagation of mouse embryonic stem cells. The Journal of Biological Chemistry,2008,283(39):26468-26476.
    [66]Huang C. Receptor-Fc fusion therapeutics, traps, and mimetibodyTM technology. Current Opinion in Biotechnology,2009,20(6):692-699.
    [67]Tong A H Y, Drees B, Nardelli G, et al. A combined experimental and computational strategy to define protein interaction networks for peptide recognition modules. Science,2002, 295(5553):321-324.
    [68]Low SC, Nunes SL, Bitonti AJ, et al. Oral and pulmonary delivery of FSH-Fc fusion proteins via neonatal Fc receptor-mediated transcytosis. Hum Reprod,2005,20(7):1805-1813.
    [69]Kitajima T, Obuse S, Adachi T, et al. Recombinant human gelatin substitute with photoreactive properties for cell culture and tissue engineering. Biotechnology arid Bioengineering,2011,108(10):2468-2476.
    [70]Gomes S, Leonor I B, Mano J F, et al. Spider silk-bone sialoprotein fusion proteins for bone tissue engineering. Soft Matter,2011,7(10):4964-4973.
    [71]Yeganegi M, Kandel RA, Santerre JP. Characterization of a biodegradable electrospun polyurethane nanofiber scaffold:Mechanical properties and cytotoxicity. Acta Biomater, 2010,6(10):3847-3855.
    [72]Tai BC, Du C, Gao S, et al. The use of a polyelectrolyte fibrous scaffold to deliver differentiated hMSCs to the liver. Biomaterials,2010,31(1):48-57.
    [73]Holzwarth J, Ma PX. Biomimetic nanofibrous scaffolds for bone tissue engineering. Biomaterials,2011,32(36):9622-9629.
    [74]Baker BM, Nathan AS, Gee AO, Mauck RL. The influence of an aligned nanofibrous topography on human mesenchymal stem cell fibrochondrogenesis. Biomaterials,2010, 31(24):6190-6200.
    [75]Yin Z, Chen X, Chen JL, et al. The regulation of tendon stem cell differentiation by the alignment of nanofibers. Biomaterials,2010,31(8):2163-2175.
    [76]Baker BM, Gee AO, Sheth NP, et al. Meniscus tissue engineering on the nanoscale:from basic principles to clinical application. J Knee Surg,2009,22(1):45-59.
    [77]Antonios G Mikos, Johnnas, Temenoff. Formation of highly porous biodegradable scaffolds for tissure engineering. Electronic Journal of Biotechnology,2000,3(2):114-119.
    [78]Mourino V, Pablo Cattalini J, Boccaccini AR. Metallic ions as therapeutic agents in tissue engineering scaffolds:an overview of their biological applications and strategies for new developments. Journal of the Royal Society Interface,2012,9(68):401-419.
    [79]Sahoo S, Ouyang H, Goh JC, et al. Characterization of a novel polymeric scaffold for potential application in tendon/ligament tissue engineering. Tissue Engineering,2006,12(1): 91-99.
    [80]Borzacchiello A, Gloria A, Mayol L, et al. Natural/synthetic porous scaffold designs and properties for fibro-cartilaginous tissue engineering. Journal of Bioactive and Compatible Polymers,2011,26(5):437-451.
    [81]Baker SC, Rohman G, Southgate J, et al. The relationship between the mechanical properties and cell behaviour on PLGA and PCL scaffolds for bladder tissue engineering. Biomaterials, 2009,30(7):1321-1328.
    [82]Hutmacher DW. Scaffolds in tissue engineering bone and cartilage. Biomaterials,2000, 21(24):2529-2543.
    [83]Werkmeister JA, Ramshaw JAM. Recombinant protein scaffolds for tissue engineering. Biomedical Materials,2012,7(1):2-12.
    [84]Gumbiner BM. Cell adhesion:the molecular basis of tissue architecture and morphogenesis. Cell,1996,84(3):345-357.
    [85]Edelman GM, Crossin KL. Cell adhesion molecules:implications for a molecular histology. Annual Review of Biochemistry,1991,60(1):155-190.
    [86]Angst BD, Marcozzi C, Magee Al. The cadherin superfamily:Diversity in form and function. Journal of Cell Science,2001,114(4):629-641.
    [87]Milner R, Campbell IL. The integrin family of cell adhesion molecules has multiple functions within the CNS. J Neurosci Res,2002,69(3):286-291.
    [88]Brummendorf T, Lemmon V. Immunoglobulin superfamily receptors:cis-interactions, intracellular adapters and alternative splicing regulate adhesion. Curr Opin Cell Biol,2001, 13(5):611-618.
    [89]Cummings RD, Smith DF. The selectin family of carbohydrate-binding proteins:structure and importance of carbohydrate ligands for cell adhesion. Bioessays,1992,14(12):849-856.
    [90]Huntley GW, Gil O, Bozdagi O. The cadherin family of cell adhesion molecules:multiple roles in synaptic plasticity. Neuroscientist,2002,8(3):221-33.
    [91]Belkin AM, Stepp MA. Integrins as receptors for laminins. Microsc Res Tech,2000,51(3): 280-301.
    [92]Crossin KL, Krushel LA. Cellular signaling by neural cell adhesion molecules of the immunoglobulin superfamily. Dev Dyn,2000,218(2):260-279.
    [93]Chou KC. The convergence-divergence duality in lectin domains of selectin family and its implications. FEBS Lett,1995,363(1-2):123-126.
    [94]Bianco P, Robey PG. Stem cells in tissue engineering. Nature,2001,414(6859):118-121.
    [95]Gojo S, Toyoda M, Umezawa A. Tissue engineering and cell-based therapy toward integrated strategy with artificial organs. Journal of Artificial Organs,2011,14(3):171-177.
    [96]Mckay R. Stem cells-hype and hope. Nature,2000,406(1):361-364.
    [97]Soler D, Gearhart J. Putting stem cells to work. Science,1999,283(5407):1468-1470.
    [98]Watt FM, Hogan BL. Out of eden:stem cells and their niches. Science,2000,287(5457): 1427-1430.
    [99]Vogel G. Harnessing the power of stem cells. Science,1999,283(5407):1432-1434.
    [100]Rippon H.J., A.E. Bishop. Embryonic stem cells. Cell Prolif,2004,37(1):23-34.
    [101]Segev H, Fishman B., Ziskind A, et al. Differentiation of human embryonic stem cells into insulin-producing clusters. Stem Cells,2004,22(3):265-274.
    [102]Kim J. Dopamine neurons derived from embryonic stem cells function in an animal model of Parkinson's disease. Nature,2002,4(418):50-56.
    [103]Cross WR, Thomas DF, Southgate J. Tissue engineering and stem cell research in urology. BJU Int,2003,92(2):165-171.
    [104]Wobus AM,Wolf E, Beier HM. Embryonic stem cells and nuclear transfer strategies. Cells Tissues Organs,2000,166(1):1-5.
    [105]Denker HW. Embryonic stem cells:an exciting field for basic research and tissue engineering, but also an ethical dilemma. Cells Tissue Organs,1999,165(324):246-249.
    [106]K. Takahashi, K. Tanabe, S. Yamanaka, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell,2007,131(5):861-872.
    [107]K. Takahashi and S. Yamanaka. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors, Cell,2006,126(4):663-676.
    [108]Hyun I, Hochedlinger K, Yamanaka S, et al. New advances in iPS cell research do not obviate the need for human embryonic stem cells. Cell stem cell,2007,1(4):367-368.
    [109]Narazaki G, Uosaki H, Yamashita JK, et al. Directed and systematic differentiation of cardiovascular cells from mouse induced pluripotent stem cells. Circulation,2008,118(5): 498-506.
    [110]Yu J, Vodyanik MA, Thomson JA, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science,2007,318(5858):1917-1920.
    [111]Ebert AD, Yu J, Svendsen CN, et al. Induced pluripotent stem cells from a spinal muscular atrophy patient. Natrue,2009,457(7227):277-280.
    [112]Song Z, Liu Y, Deng H, et al. Efficient generation of hepatocyte-like cells from human induced pluripotent stem cells. Cell research,2009,19(11):1233-1242.
    [113]John T, Kit T, Kevin Eggan, et al. Induced pluripotent stem cells generated from patients with ALS can be differentiated into motor neurons. Science,2008,321(5893):1218-1221.
    [114]Hodgkinson T, Yuan X F, Bayat A. Adult stem cells in tissue engineering. Expert Review of Medical Devices,2009,6(6):621-640.
    [115]Presnell S.C., B. Petersen, M. Heidaran. Stem cells in adult tissues. Cell & Dev. Biol,2002, (13):369-376.
    [116]Eberli D, Atala A. Tissue engineering using adult stem cells, in:Klimanskaya ILR, eds. Stem Cell Tools and Other Experimental Protocols.2006:287-302.
    [117]Fan CG, Zhang QJ, Zhou JR. Therapeutic potentials of mesenchymal stem cells derived from human umbilical cord. Stem Cell Reviews and Reports,2011,7(1):195-207.
    [118]Guilak F, Awad HA, Fermor B, et al. Adipose-derived adult stem cells for cartilage tissue engineering. Biorheology,2004,41(3-4):389-399.
    [119]Caplan AI. Adult mesenchymal stem cells for tissue engineering versus regenerative medicine. Journal of Cellular Physiology,2007,213(2):341-347.
    [120]Koc, ON, Lazarus HM. Mesenchymal stem cells:heading into the clinic. Bone Marrow Transplantation,2001,27(12):235-239.
    [121]Tekkatte C, Vidyasekar P, Kapadia NK, et al. Enhancement of adipogenic and osteogenic differentiation of human bone-marrow-derived mesenchymal stem cells by supplementation with umbilical cord blood serum. Cell and Tissue Research,2012,347(2):383-395.
    [122]Chamberlain G, Fox J, Ashton B, et al. Concise review:Mesenchymal stem cells:Their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells,2007,25(11):2739-2749.
    [123]Jung S, Panchalingam KM, Rosenberg L, et al. Ex vivo expansion of human mesenchymal stem cells in defined serum-free media. Stem Cells International,2012,2012(1):1-2.
    [124]Patel SA, Sherman L, Munoz J, et al. Immunological properties of mesenchymal stem cells and clinical implications. Archivum Immunologiae et Therapiae Experimentalis,2008,56(1): 1-8.
    [125]Seib FP, Miiller K, Franke M, et al. Engineered extracellular matrices modulate the expression profile and feeder properties of bone marrow-derived human multipotent mesenchymal stromal cells. Tissue Engineering Part A,2009,15(10):3161-3171.
    [126]Prewitz M, Seib FP, Pompe T, et al. Polymeric biomaterials for stem cell bioengineering. Macromolecular Rapid Communications,2012,33(17):1420-1431.
    [127]Sakai S, Kim J, Hexig B, et al. Adsorption behaviors of recombinant E-cadherin-IgG Fc fusion protein on polystyrene surface. Colloids and Surfaces B:Biointerfaces,2012,94(1): 192-198.
    [128]Azuma K, Nagaoka M, Cho C-S, et al. An artificial extracellular matrix created by hepatocyte growth factor fused to IgG-Fc. Biomaterials,2010,31(5):802-809.
    [129]Yu M, Du F, Ise H, et al. Preparation and characterization of a VEGF-Fc fusion protein matrix for enhancing HUVEC growth. Biotechnology Letters,2012,34(9):1765-1771.
    [130]Song G, Ju Y, Shen X, et al. Mechanical stretch promotes proliferation of rat bone marrow mesenchymal stem cells. Colloids and Surfaces B:Biointerfaces,2007,58(2):271-277.
    [131]Roy F, Berx G. The cell-cell adhesion molecule E-cadherin. Cellular and Molecular Life Sciences,2008,65(23):3756-3788.
    [132]Yonemura S. Cadherin-actin interactions at adherens junctions. Current Opinion in Cell Biology,2011,23(5):515-522.
    [133]Delva E, Kowalczyk AP. Regulation of cadherin trafficking. Traffic,2009,10(3):259-267.
    [134]Semler EJ, Dasgupta A, Moghe PV. Cytomimetic engineering of hepatocyte morphogenesis and function by substrate-based presentation of acellular e-cadherin. Tissue engineering, 2005,11(5-6):734-750.
    [135]Li L, Bennett SAL, Wang L. Role of E-cadherin and other cell adhesion molecules in survival and differentiation of human pluripotent stem cells. Cell Adhesion & Migration, 2012,6(1):59-70.
    [136]Stockinger A. E-cadherin regulates cell growth by modulating proliferation-dependent beta-catenin transcriptional activity. The Journal of Cell Biology,2001,154(6):1185-1196.
    [137]Brieva TA, Moghe PV. Exogenous cadherin microdisplay can interfere with endogenous signaling and reprogram gene expression in cultured hepatocytes. Biotechnology and Bioengineering,2004,85(3):283-292.
    [138]Kelly KF, Ng DY, Jayakumaran G, et al.β-catenin enhances oct-4 activity and reinforces pluripotency through a tcf-independent mechanism. Cell Stem Cell,2011,8(2):214-227.
    [139]Nelson WJ. Regulation of cell-cell adhesion by the cadherin-catenin complex. Biochemical Society Transactions,2008,36(2):149.
    [140]Tom K. Kuo, Shun-pei Hung, Oscar K. Lee, et al. Stem cell therapy for liver disease: parameters governing the success of using bone marrow mesenchymal stem cells. Gastroenterology,2008,134(7):2111-2121.
    [141]Horwitz EM. Stem cell plasticity:the growing potential of cellular therapy. Archives of Medical Research,2003,34(6):600-606.
    [142]Chen Y, Dong XJ, Zhang GR, et al. In vitro differentiation of mouse bone marrow stromal stem cells into hepatocytes induced by conditioned culture medium of hepatocytes. Journal of Cellular Biochemistry,2007,102(1):52-63.
    [143]Takenaga M, Fukumoto M, Hori Y. Regulated nodal signaling promotes differentiation of the definitive endoderm and mesoderm from ES cells. Journal of Cell Science,2007,120(12): 2078-2090.
    [144]Jianbin Xu, Chuanshun Zhu, Jun Yang, et al. hE-cadherin-Fc fusion protein coated surface enhances the adhesionand proliferation of human mesenchymal stem cells. Colloids and Surfaces B:Biointerfaces,2013,109(1):97-102.
    [145]Farshid Guilak, Daniel M. Cohen, Bradley T. Estes, et al. Control of stem cell fate by physical interactions with the extracellular matrix. Cell Stem Cell,2009,5(1):17-26.
    [146]Silvia S. Chen, Wendy Fitzgerald, Joshua Zimmerberg. Cell-cell and cell-extracellular matrix interactions regulate embryonic stem cell differentiation. Stem Cells,2007,25(3): 553-561.
    [147]A. Steplewski, T. Alabyeva, A. Gertala. Testing the utility of rationally engineered recombinant collagen-like proteins for applications in tissue engineering. Journal of Biomedical Materials Research Part A,2006,76(3):551-560.
    [148]Hersel. U, C. Dahmen, H. Kessler. RGD modified polymers:biomaterials for stimulated cell adhesion and beyond. Biomaterials,2003,24(24):4385-4415.
    [149]Nikolaos T. Georgopoulos, Lisa A. Kirkwood, Dawn C. Walker, et al. Differential regulation of growth-promoting signalling pathways by E-cadherin. PLoS ONE,2010,5(10): 13621-13634.
    [150]Alistair Watt, Wendyd Garrison, Stephena Duncan, et al. HNF4:A central regulator of hepatocyte differentiation and function. Hepatology,2003,37(6):1249-1253.
    [151]Hans Clevers. Wnt/β-catenin signaling in development and disease. Cell,2006,127(3): 469-480.
    [152]Sell S, Barnes C, Smith M, et al. Extracellular matrix regenerated:Tissue engineering via electrospun biomimetic nanofibers. Polymer International,2007,56(11):1349-1360.
    [153]Dalby MJ, Gadegaard N, Tare R, et al. The control of human mesenchymal cell differentiation using nanoscale symmetry and disorder. Nature Materials,2007,6(12): 997-1003.
    [154]Hollister S, Maddox R, Taboas J. Optimal design and fabrication of scaffolds to mimic tissue properties and satisfy biological constraints. Biomaterials,2002,23(20):4095-4103.
    [155]Badylak S, Freytes D, Gilbert T. Extracellular matrix as a biological scaffold material: Structure and function. Acta Biomaterialia,2009,5(1):1-13.
    [156]Venugopal J, Low S, Choon AT, et al. Interaction of cells and nanofiber scaffolds in tissue engineering. Journal of Biomedical Materials Research Part B:Applied Biomaterials,2007, 84(1):34-48.
    [157]Ma Z, Kotaki M, Inai R, et al. Potential of nanofiber matrix as tissue-engineering scaffolds. Tissue engineering,2005,11(1-2):101-109.
    [158]Boudriot U, Dersch R, Greiner A, et al. Electrospinning approaches toward scaffold engineering-a brief overview. Artificial organs,2006,30(10):785-792.
    [159]Pham QP, Sharma U, Mikos AG. Electrospinning of polymeric nanofibers for tissue engineering applications:A review. Tissue engineering,2006,12(5):1197-1211.
    [160]Hussain F. Review article:Polymer-matrix nanocomposites, processing, manufacturing, and application:An overview. Journal of Composite Materials,2006,40(17):1511-1575.
    [161]Han D, Gouma P-I. Electrospun bioscaffolds that mimic the topology of extracellular matrix. Nanomedicine:Nanotechnology, Biology and Medicine,2006,2(1):37-41.
    [162]Pompe T, Keller K, Mothes G, et al. Surface modification of poly(hydroxybutyrate) films to control cell-matrix adhesion. Biomaterials,2007,28(1):28-37.
    [163]Haque MA, Nagaoka M, Hexig B, et al. Artificial extracellular matrix for embryonic stem cell cultures:A new frontier of nanobiomaterials. Science and Technology of Advanced Materials,2010,11(1):014106.
    [164]Lannutti J, Reneker D, Ma T, et al. Electrospinning for tissue engineering scaffolds. Materials Science and Engineering:C,2007,27(3):504-509.
    [165]Du F, Zhao W, Zhang M, et al. The synergistic effect of aligned nanofibers and hyaluronic acid modification on endothelial cell behavior for vascular tissue engineering. Journal of Nanoscience and Nanotechnology,2011,11(8):6718-6725.
    [166]Li WJ, Danielson KG, Alexander PG, et al. Biological response of chondrocytes cultured in three-dimensional nanofibrous poly (□-caprolactone) scaffolds. Journal of Biomedical Materials Research Part A,2003,67(4):1105-1114.
    [167]Jin H. Human bone marrow stromal cell responses on electrospun silk fibroin mats. Biomaterials,2004,25(6):1039-1047.
    [168]Li WJ, Tuli R, Huang X, et al. Multilineage differentiation of human mesenchymal stem cells in a three-dimensional nanofibrous scaffold. Biomaterials,2005,26(25):5158-5166.
    [169]Lu LX, Wang YY, Mao X, et al. The effects of PHBV electrospun fibers with different diameters and orientations on growth behavior of bone-marrow-derived mesenchymal stem cells. Biomedical Materials,2012,7(1):015002.
    [170]Williams DF. To engineer is to create:The link between engineering and regeneration. Trends Biotechnol,2006,24(1):4-8.
    [171]Engelberg I, Kohn J. Physico-mechanical properties of degradable polymers used in medical applications:A comparative study. Biomaterials,1991,12(3):292-304.
    [172]Ueda H, Tabata Y. Polyhydroxyalkanonate derivatives in current clinical applications and trials. Adv Drug Deliv Rev,2003,55(4):501-518.
    [173]Tan PS, Teoh SH. Effect of stiffness of polycaprolactone (PCL) membrane on cell proliferation. Mater Sci Eng C,2007,27(2):304-308.
    [174]Tang ZG, Hunt JA. The effect of PLGA doping of polycaprolactone films on the control of osteoblast adhesion and proliferation in vitro. Biomaterials,2006,27(25):4409-4418.
    [175]Weigel T, Schinkel G, Lendlein A. Design and preparation of polymeric scaffolds for tissue engineering. Expert Rev Med Dev,2006,3(6):835-851.
    [176]Leong KF, Cheah CM, Chua CK. Solid freeform fabrication of three-dimensional scaffolds for engineering replacement tissues and organs. Biomaterials,2003,24(13):2363-2378.
    [177]Chen GP, Ushida T, Tateishi T. Scaffold design for tissue engineering. Macromol Biosci, 2002,2(2):67-77.
    [178]Mikos AG, Temenoff JS. Formation of highly porous biodegradable scaffolds for tissue engineering. Elec J Biotech,2000,3(2):114-119.
    [179]Sachlos E, Czernuszka JT. Making tissue engineering scaffolds work. Review:the application of solid freeform fabrication technology to the production of tissue engineering scaffolds. Eur Cell Mater,2003,5(30):29-40.
    [180]Baker SC, Atkin N, Gunning PA, et al. Characterisation of electrospun polystyrene scaffolds for three-dimensional in vitro biological studies. Biomaterials,2006,27(16):3136-3146.
    [181]Griffith LG. Polymeric biomaterials. Acta Mater,2000,48(15):263-277.
    [182]Sung HJ, Meredith C, Johnson C, et al. The effect of scaffold degradation rate on three-dimensional cell growth and angiogenesis. Biomaterials,2004,25(26):5735-5742.

© 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