PEG交联云细胞瓣多信号复合材料生物学性能研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
第一部分:PEG交联去细胞瓣复合材料构建及其体外生物相容性
     目的:PEG交联去细胞瓣构建复合材料,并评价其体外生物相容性。
     方法:(1)合成功能团为丙烯酰基的枝化状PEG,同时在去细胞瓣上引入巯基,通过丙烯酰基与巯基发生迈克尔加成反应,构建PEG交联去细胞瓣复合材料。(2)生物力学测试评价PEG交联对去细胞瓣机械性能的影响;CCK-8法检测复合材料浸提液对人脐静脉内皮细胞增殖率影响,以评价其生物相容性。
     结果:复合材料构建条件温和、效果确切;复合材料抗拉强度明显高于去细胞瓣(P<0.05),且与天然瓣膜抗拉强度相比,差异无统计学差异(P>0.05)。复合材料与去细胞瓣毒性评级均为0级,与阴性对照组相比,差异无统计学意义(P>0.05);人脐静脉内皮细胞在含复合材料浸提液的培养液中形态良好,增殖旺盛。
     结论:PEG交联去细胞瓣的方法能够构建与天然瓣膜相似抗拉强度且无细胞毒性的复合材料。
     第二部分:PEG交联去细胞瓣多信号复合材料构建
     第一节:构建RGD-复合材料
     目的:评价PEG交联去细胞瓣复合材料结合粘附肽(精-甘-天冬氨酸RGD肽)的可行性和有效性。
     方法:复合材料与1ml不同浓度(1.5mg/ml, 1mg/ml,0.5mg/ml,0.25mg/ml)GRGDSPC肽溶液在37℃反应,在不同时间点(1h,2h,4h),用分光光度法检测复合材料结合GRGDSPC肽质量,并行免疫荧光定性检测,去细胞瓣作为阴性对照。
     结果:GRGDSPC肽能有效地结合于复合材料,且一片复合材料与lml GRGDSPC肽(1mg/ml)在37℃条件下反应2h,结合GRGDSPC肽质量最大为(0.79±0.01)mg。
     结论:PEG交联去细胞瓣复合材料,能够在体外有效地结合RGD肽,构建RGD-复合材料。
     第二节:构建VEGF-复合材料
     目的:评价PEG交联去细胞瓣复合材料结合血管内皮细胞生长因子(VEGF)的可行性和有效性。
     方法:复合材料与1ml不同浓度(500pg/ml, 1000pg/ml,2000pg/ml) VEGF溶液在37℃反应,在不同时间点(1h,2h,4h,12h),用ELISA法检测复合材料结合VEGF质量,并行免疫荧光检测,去细胞瓣作为阴性对照。
     结果:VEGF能有效地结合于复合材料,且一片复合材料与1ml VEGF (1000pg/ml)在37℃反应4h,其结合VEGF质量最大为(896.87+3.27)pg。
     结论:PEG交联去细胞瓣复合材料,能够在体外有效地结合VEGF,构建VEGF-复合材料。
     第三节:构建PEG交联去细胞瓣多信号复合材料
     目的:评价PEG交联去细胞瓣复合材料同时结合RGD肽和VEGF的可行性和有效性。
     方法:复合材料与1ml反应液(包含1mg/mlGRGDSPC肽和1000pg/mlVEGF)在37℃反应4h,对反应后复合材料行免疫荧光检测,去细胞瓣作为阴性对照。
     结果:GRGDSPC肽和VEGF能同时有效地结合于复合材料。
     结论:PEG交联去细胞瓣复合材料,能够在体外有效地结合RGD肽和VEGF,构建PEG交联去细胞瓣多信号(RGD、VEGF)复合材料。
     第三部分:PEG交联去细胞瓣多信号复合材料生物学性能研究
     第一节:内皮祖细胞分离、培养与鉴定
     目的:分离、培养、鉴定脐带血内皮祖细胞,为其作为组织工程种子细胞提供实验依据。
     方法:密度梯度离心法分离新鲜脐血中单个核细胞,在含碱性成纤维细胞生长因子和血管内皮生长因子培养液中培养,通过形态学、免疫荧光和流式细胞仪等对贴壁细胞进行鉴定;并与脐静脉内皮细胞进行增殖和迁移能力比较。
     结果:随培养时间延长,贴壁细胞形态发生明显改变,从小圆变成梭形,逐渐形成特征性克隆和典型成熟内皮细胞鹅卵石样形态;体外培养7天后,90%以上贴壁细胞呈Dil-ac-LDL和FITC-UEA-Ⅰ双染阳性;贴壁细胞流式细胞仪分析显示:培养7 d的细胞VEGFR-2、CD34和CD133表达分别占(77.35+4.86)%、(52.42+6.64)%和(19.36+2.14)%,培养28天的细胞VEGFR-2和CD34表达分别占(81.06+7.31)%和(7.62±3.14)%,而未检测到CD133表达;人内皮祖细胞增殖和迁移能力明显高于人脐静脉内皮细胞<0.05)。
     结论:用密度梯度离心法和贴壁筛选法,可成功分离出脐带血内皮祖细胞,该方法简单、经济、可行;且内皮祖细胞可向内皮细胞分化,增殖和迁移能力强,有望成为理想种子细胞。
     第二节:PEG交联去细胞瓣多信号复合材料生物学性能测定
     目的:研究PEG交联去细胞瓣多信号(RGD、VEGF)复合材料对内皮祖细胞粘附和增殖的影响,为进一步构建TEHV提供依据。
     方法:A组去细胞瓣,B组PEG交联去细胞瓣复合材料,C组VEGF-复合材料,D组为RGD-复合材料,E组PEG交联去细胞瓣多信号(RGD、VEGF)复合材料。然后在各组材料上种植EPCs,分别在种植后2h、4h、8h用细胞计数和3H-TdR掺入法检测各组材料上细胞数和每分钟脉冲数,反映EPCs在各组材料上粘附性;分别在种植后2d、4d、8d,用细胞计数和3H-TdR掺入法检测各组瓣膜支架上细胞数和每分钟脉冲数,反映各组材料上EPCs增殖情况。在种植8d时,取各组瓣膜支架行苏木素-伊红染色和扫描电镜,以检测EPCs在各组材料上生长情况,并取E组支架材料上细胞行RT-PCR检测t-PA和eNOS的表达,以评价新内皮抗血栓形成功能。
     结果:(1)种植后2h、4h、8h时各组细胞数和每分钟脉冲数为:D组>E组>C组>A组>B组,其中A组与B组之间各个时间点均无统计学差异(P>0.05),8h时D组与E组之间的差异无统计学意义(P>0.05),其余时间点各组之间的差异均有统计学意义(P<0.05)。(2)种植2d、4d、8d时各组细胞数和每分钟脉冲数为:A组均大于B组(P>0.05),C组、D组和E组均大于同一时间点A组和B组(P<0.05);种植2d时,E组>C组(P<0.05),D组>E组(P>0.05);种植4d时,E组>D组(P<0.05),D组>C组(P<0.05);种植8d时,E组>D组(P<0.05),D组>C组(P>0.05)。(3)苏木素-伊红染色和扫描电镜检测显示:种植8d后,与其它组相比,E组瓣膜支架上可见一致密融合细胞层,并且该新内皮层细胞t-PA和eNOS表达与人脐静脉内皮细胞相似。
     结论:PEG交联去细胞瓣多信号(RGD、VEGF)复合材料上RGD肽和VEGF,能够协同促进内皮祖细胞在复合材料上粘附和增殖,有利于TEHV构建。
Part one:Construction and in vitro biocompatibility of the composite PEG-crosslinked decellularized valve
     Objective:To construct a composite material by polyethylene glycol (PEG) crosslinking of decellularized valves, and evaluate the biocompatibility of the composite material in vitro.
     Methods:First, branched PEG with the functional group of propylene acyl was synthesized, and thiol was introduced into decellularized valves. Then the composite valve was constructed by the Michael addition reaction between propylene acyl and thiol. Second, the effects of PEG crosslinking on biomechanical properties of decellularized valve were evaluated. For assessing the biocompatibility, cell proliferation rates of the human umbilical vein endothelial cells (HUVECs) which were exposed to the extract of composite material were detected by CCK-8 assay.
     Results:The PEG-crosslinking reaction for the construction of composite material has a mild conditions and a high efficiency; The tensile strength of the composite materials was obviously higher than that of the decellularized ones(P<0.05), and there was no significant difference in tensile strength between composite materials and natural valves(P>0.05). The toxicity of composite material were classified Grade 0, and no significant difference of toxicity was observed between the composite material group and the negative control group; HUVECs treated with culture medium containing extracts of composite material show great cytoactivity and normal morphology.
     Conclusions:A noncytotoxic composite material can be obtained by PEG crosslinking with the decellularized valves, and the tensile strength of the composite material is similar to the natural one.
     Part two:Construction of the composite PEG-crosslinked decellularized valve with multi-signal
     Chapter one:Construction of the composite valve conjugated with RGD peptide
     Objective:To evaluate the feasibility and effectiveness of conjugation of RGD (Arg-Gly-Asp) peptide on the composite PEG-crosslinked decellularized valve and optimize the reaction condition.
     Methods:The composite PEG-crosslinked decellularized valve was reacted with 1ml GRGDSPC peptides solution of different concentrations (1.5mg/ml, 1mg/ml, 0.5mg/ml,0.25mg/ml) at 37℃. The conjugated effect was evaluated quantitatively by spectrophotometer at different time points (1h,2h, 4h), and the qualitative tests were performed by immunofluorescence. The decellularized valves were set as negative control.
     Results:The GRGDSPC peptides could be conjugated effectively with the PEG-composite valve, and the quality of conjugated GRGDSPC peptides reaches a climax amount of (0.79±0.01)mg when the concentration of GRGDSPC peptides solution was lmg/ml and the reaction time was 2h.
     Conclusion:The construction of RGD-composite material can be achieved by grafting GRGDSPC peptide on the composite PEG-crosslinked decellularized valve in vitro.
     Chapter two Construction of the composite valve conjugated with VEGF
     Objective:To evaluate the feasibility and effectiveness of conjugation of VEGF on composite PEG-crosslinked decellularized valve and optimize the reaction condition.
     Methods:The composite PEG-crosslinked decellularized valve was reacted with lml VEGF solution of different concentrations (500pg/ml, 1000pg/ml,2000pg/ml) at 37℃. The quality of conjugated VEGF was measured by ELISA at different time points (1h,2h,4h,12h), and the qualitative tests were performed by immunofluorescence. The decellularized valves were set as negative control.
     Results:The VEGF could be conjugated effectively on the composite valve by PEG, and the maximum quality of conjugated VEGF was (896.87±3.27)pg when the concentration of VEGF solution was 1000pg/ml and reaction time was 4h.
     Conclusion:The construction of VEGF-composite material can be achieved by grafting VEGF on the composite PEG-crosslinked decellularized valve in vitro.
     Chapter three:Construction of the composite PEG-crosslinked decellularized valve with multi-signal
     Objective:To assess the probability and effectiveness of conjugation of RGD peptide and VEGF on composite PEG-crosslinked decellularized valve simultaneously.
     Methods:The composite PEG-crosslinked decellularized valve were reacted with lml reaction mixture solution (including lmg/ml GRGDSPC peptide and 1000pg/ml VEGF) at 37℃for 4h. Then the immunofluorescence was taken to make a qualitative evaluation of the conjugation effects. The decellularized valves were set as negative control.
     Results:The GRGDSPC peptides and VEGF could be conjugated effectively onto composite material by PEG.
     Conclusion:The construction of composite PEG-crosslinked decellularized valve with multi-signal (RGD and VEGF) can be achieved by conjugating the RGD peptide and VEGF on the composite PEG-crosslinked decellularized valve in vitro.
     Part three:Study on biological properties of the composite PEG-crosslinked decellularized valve with multi-signal
     Chapter one:The isolation, Culture and identification of EPCs
     Objective:To provide human endothelial progenitor cells (EPCs) for the further study by isolation, culture and identification of EPCs from human umbilical cord blood.
     Methods:Fresh umbilical blood was collected and density gradient centrifugated to isolate mononuclear cells (MNCs). The cells then were cultured in the medium supplemented with VEGF and bFGF. Morphology, immunofluorescence and flow cytometry were applied to confirm that the attached cells were EPCs. The proliferation and migration capability of EPCs were compared with HUVECs.
     Results:As the time went on, the shape of the attached cells changed from small round to spindle. the typical colonies were gradually formed and the cobblestone-like morphology as a specific characteristic of mature endothelial cells was observed; after been cultured for 7 days, more than 90% of the attached cells express both Dil-acLDL and FITC-UEA-Ⅰ. Flow cytometric analysis showed that the positive staining rate of the attached cells for VEGFR2、CD34 and CD133 were (77.35±4.86)%, (52.42±6.64)% and (19.36±2.14)% respectively, and at the 28th day, the positive staining rate of attached cells for VEGFR2 and CD34 were (81.06±7.31)% and (7.62±3.14)% respectively while CD133 can not been stained. Compared with HUVECs, EPCs have more potent potential of proliferation and migration (P<0.05).
     Conclusion:EPCs can be isolated successfully from human umbilical cord blood by a simple, economical and feasible method composed of density gradient centrifugation and adherent filtration. EPCs can be introduced to endothelial cells and have great capacity of proliferation and migration, which makes it an ideal seed cells.
     Chapter two:Measurement on biological properties of the composite PEG-crosslinked decellularized valve with multi-signal
     Objective:To investigate the effect of composite valve with multi-signal on the adhesion and proliferation of EPCs, and provide the basis for the further construction of TEHV
     Methods:The experiment were assigned into 5 groups:Group A were decellularized valves, Group B were composite PEG-crosslinked decellularized valves, Group C were VEGF-composite valve, Group D were RGD-composite valve and Group E were multi-signal composite valve(n=8). EPCs were seeded onto the valve of different groups, the cells number and counts per minute(cpm) were detected by cell counting and thymidine(3H-TdR) up-take method at 2h、4h、8h respectively after seeding to estimate the adhesion amount of EPCs in different groups. the cells number and cpm of each group were detected using cell counting and thymidine(3H-TdR) up-take method at 2d、4d、8d respectively after seeding to evaluate the proliferation of EPCs. Hematoxylin and eosin (HE) stain and scanning electron microscopy (SEM) were performed to give a morphology evidence of the growth of EPCs at 8d after seeding. Finally the expression of t-PA and eNOS of group E were measured by RT-PCR to assess the antithrombotic function of the multi-signal composite valve.
     Results:(1) After been seeded for 2h、4h and 8h, the results of cell count and cpm in each group were as the following:Group D>Group E>Group C>Group A>Group B, but there was no significant difference between Group A and Group B(P> 0.05), there was also no significant difference between Group D and Group E at the 8h after the seeding(P>0.05), but the differences in the other groups were significant at the other time points(P<0.05), (2) At the 2d,4d and 8d after been seeded, the results of cell count and cpm in each group were as the following: Group A was always higher than Group B(P>0.05), the cell number and cpm in group C、Group D and Group E were significantly higher than those in Group A and Group B (P<0.05) at the time point. At the 2d after the seeding, the cell number and cpm in Group E was higher than those in Group C (P<0.05), the cell number and cpm in Group D was higher than those in Group E(P>0.05); at the 4d after the seeding, the cell number and cpm in Group E higher those in group D(P<0.05), the cell number and cpm in Group D was higher than those in Group C(P<0.05); at the 8d after the seeding, the cell number and cpm in Group E was higher than those in Group D(P<0.05), the cell number and cpm in Group D was higher than those in Group C(P>0.05). (3) the results of HE and SEM indicated that the confluent and compact monolayer could be formed on the surface of Group E compared with other groups at 8d after the seeding, and the expression level of t-PA and eNOS gene in this neo-endothelium were very similar to those in HUVECs.
     Conclusion:The RGD peptides and VEGF conjugated on the composite PEG-crosslinked decellularized valve can achieve a synergistic promotion on the adhesion and proliferation of EPCs seeded on the composite valve, which is useful for the construction of tissue engineering heart valves.
引文
[1]. Nkomo VT, Gardin JM, Skelton TN, et al. Burden of valvular heart diseases:a population-based study. Lancet,2006,368(9540):1005-1011.
    [2]. Rosamond W, Flegal K, Furie K, et al. Heart disease and stroke statistics-2008 update: a report from the American Heart Association statistics committee and stroke statistics subcommittee. Circulation,2008,117:E25-E146.
    [3]. Yacoub MH, Takkenberg JJM. Will heart valve tissue engineering change the world? Nat Clin Pract Cardiovasc Med,2005,2(2):60-61.
    [4]. Mikos AG, Herring SW, Ochareon P, et al. Engineering complex tissues. Tissue Eng, 2006,12(12):3307-3339.
    [5]. Mayer JE Jr. Uses of homograft conduits for right ventricle to pulmonary artery connections in the neonatal period. Semin Thorac Cardiovasc Surg,1995,7(3): 130-2.
    [6]. Schoen FJ, Levy RJ. Founder's Award,25th Annual Meeting of the Society for Biomaterials, perspectives. Providence, RI, April 28-May 2,1999. Tissue heart valves: current challenges and future research perspectives. J Biomed Mater Res,1999,47(4): 439-465.
    [7].董念国,史嘉玮,胡平.组织工程心脏瓣膜支架的研究进展.中华实验外科杂志,2007,24(3):261-262.
    [8]. Hammermeister KE, Sethi GK, Henderson WG, et al. A comparison of outcomes in men 11 years after heart-valve replacement with a mechanical valve or bioprosthesis.Veterans Affairs Cooperative Study on Valvular Heart Disease. N Engl J Med,1993,328(18):1289-96.
    [9]. Schmidt CE, Baier JM. Acellular vascular tissues:natural biomaterials for tissue repair and tissue engineering. Biomaterials,2000,21(22):2215-2231.
    [10]. Shinoka T, Breuer CK, Tanel RE, et al. Tissue engineering heart valves:valve leaflet replacement study in a lamb model. Ann Thorac Surg,1995,60(Suppl 3):S513-S516.
    [11]. Langer R, Vacanti JP. Tissue engineering. Science,1993,260:920-6.
    [12]. Vacanti JP, Langer R. Tissue engineering:the design and fabrication of living replacement devices for surgical reconstruction and transplantation. Lancet,1999, 354(Suppl 1):SI32-4.
    [13]. Hong H, Dong GN, Shi WJ, et al. Fabrication of biomatrix/polymer hybrid scaffold for heart valve tissue engineering in vitro. ASAIO J,2008,54(6):627-632.
    [14]. Mendelson K, Schoen FJ. Heart valve tissue engineering:concepts, approaches, progress, and challenges. Ann Biomed Eng,2006,34(12):1799-1819.
    [15]. Kidane AG, Burriesci G, Cornejo P, et al. Current developments and future prospects for heart valve replacement therapy. J Biomed Mater Res B Appl Biomater,2009, 88B(1):290-303.
    [16]. Concha M, Aranda PJ, Casares CM, et al. The Ross Procedure. Journal of Cardiovascular Surgery,2004,19(4):401-409.
    [17]. Harken DE, Taylor WJ, Le Femine AA, et al. Aortic valve replacement with a caged ball valve. Am J Cardiol,1962,9:292-299.
    [18]. Bader A, Schilling T, Teebken OE, et al. Tissue engineering of heart valves-human endothelial cell seeding of detergent acellularized porcine valves. Eur J Cardiothorac surg,1999,14:279-284.
    [19]. Booth C, Korossis SA, Wilcox HE, et al. Tissue engineering of cardiac valve prostheses I:development and histological characterization of an acellular porcine scaffold. J Heart Valve Dis,2002,11(4):457-462.
    [20]. Schenke-Layland K, Vasilevski O, Opitz F, et al. Impact of decellularization of xenogeneic tissue on extracellular matrix integrity for tissue engineering of heart valves. Journal of Structural Biology,2003,143(3):201-208.
    [21].董念国,叶晓峰,孙宗全,等.脱细胞天然支架生物力学性能变化及预处理改善组织相容性的实验研究.中华外科杂志,2007,45(16):1128-1131.
    [22]. Liao J, Joyce EM, Sacks MS. Effects of decellularization on the mechanical and structural properties of the porcine aortic valve leaflet. Biomaterials,2008,29(8): 1065-1074.
    [23]. Fuchs JR, Nasseri BA, Vacanti JP. Tissue engineering:a 21st century solution to surgical reconstruction. Ann Thorac Surg,2001,72(2):577-591.
    [24]. Hu Y, Winn SR, Krajbich I, et al. Porous polymer scaffolds surface-modified with arginine-glycine-aspartic acid enhance bone cell attachment and differentiation in vitro. J Biomed Mater Res,2003,64A(3):583-590.
    [25]. Zisch AH, Lutolf MP, Ehrbar M, et al. Cell-demanded release of VEGF from synthetic, biointeractive cell ingrowth matrices for vascularized tissue growth. The FASEB Journal,2003,17:2260-2262.
    [26]. Ferrara N, Houck K, Jakeman L, et al. Molecular and biological properties of the vascular endothelial growth factor family of proteins. Endocrine Reviews,1992,13 (1):18-32.
    [27]. Kasimir MT, Weigel G, Sharma J, et al. The decellularized porcine heart valve matrix in tissue engineering:platelet adhesion and activation. Thromb Haemost,2005,94: 562-567.
    [28]. Dohmen PM, Ozaki S, Nitsch R, et al. A tissue engineered heart valve implanted in a juvenile sheep model. Med Sci Monit,2003,9(4):BR97-BR104.
    [29]. Breuer CK, Mettler BA, Anthony T, et al. Application of tissue-engineering principles toward the development of a semilunar heart valve substitute. Tissue Eng,2004, 10(11-12):1725-1736.
    [30]. Kasimir MT, Rieder E, Seebacher G, et al. Decellularization does not eliminate thrombogenicity and inflammatory stimulation in tissue-engineered porcine heart valves. J Heart Valve Dis,2006,15(2):278-286.
    [31]. Dohmen PM, Lembcke A, Hotz H, et al. Ross operation with a tissue-engineered heart valve. Ann Thorac Surg,2002,74:1438-1442.
    [32]. Asahara T, Murohara T, Sullivan A, et al. Isolation of putative progenitor endothelial cells for angiogenesis. Science,1997,275(5302):964-967.
    [33]. Wu X, Rabkin-Aikawa E, Guleserian KJ, et al. Tissue-engineered micro vessels on three-dimensional biodegradable scaffolds using human endothelial progenitor cells. Am J Physiol Heart Circ Physiol,2004,287:H480-H487.
    [34]. Hristov M, Erl W, Weber PC. Endothelial progenitor cells:isolation and characterization. Trends Cardiovasc Med,2003,13:201-206.
    [35]. Schmidt D, Breymann C, Weber A, et al. Umbilical cord blood derived endothelial progenitor cells for tissue engineering of vascular grafts. Ann Thorac Surg,2004,78: 2094-2098.
    [36]. Schmidt D, Mol A, Neuenschwander S, et al. Living patches engineered from human umbilical cord derived fibroblasts and endothelial progenitor cells. Eur J Cardiothorac Surg,2005,27(5):795-800.
    [37]. Kaushal S, Amiel GE, Guleserian KJ, et al. Functional small diameter neovessels created using endothelial progenitor cells expanded in vivo. Nat Med,2001,7(9): 1035-1040.
    [1]. Mendelson K, Schoen FJ. Heart valve tissue engineering:concepts, approaches, progress, and challenges. Ann Biomed Eng,2006,34(12):1799-1819.
    [2].洪昊,董念国.组织工程心脏瓣膜的研究进展.临床心血管病杂志,2006,22(7):442-444.
    [3].董念国,叶晓峰,史嘉玮,等.组织工程瓣膜天然支架去细胞方法的比较.中华实验外科杂志,2005,22(3):377-377.
    [4]. Shi JW, Dong NG, Sun ZQ. Immobilization of decellularized valve scaffolds with Arg-Gly-Asp-containing peptide to promote myofibroblast adhesion. J Huazhong Univ Sci Technol [Med Sci],2009,29(4):503-507.
    [5].董念国,卢翠芬,史嘉玮,等.枝化状聚乙二醇及其制备方法和用途.中国专利.No.2010101185647.
    [6]. Chen JS, Noah EM, Pallua N, et al. The use of bifunctional polyethyleneglycol derivatives for coupling of proteins to and cross-linking of collagen matrices. Journal of materials science:materials in medicine,2002,13(11):1029-1035.
    [7].邓诚,董念国,史嘉玮,等.聚乙二醇载药缓释微球复合去细胞瓣制备组织工程心脏瓣膜支架.临床心血管病杂志,2010,26(1):68-71.
    [8]. Wu S, Liu YL, Cui B, et al. Study on decellularized porcine aortic valve/poly(3-hydr-oxybutyrate-co-3-hydroxyhexanoate) hybrid heart valve in sheep model. Artificial Organs,2007,31(9):689-697.
    [9]. Baudin B, Bruneel A, Bosselut N, et al. A protocol for isolation and culture of human umbilical vein endothelial cells. Nat Protoc,2007,2(3):481-485.
    [10]. ISO 10993-12:2002, Biological evaluation of medical devices-Part 12:Sample preparation and reference materials.
    [11].郑丽君,虞渝生,刘友山,等.国产膨体聚四氟乙烯的预处理及其细胞毒性评价.材料科学与工程学报,2008,26(4):654-656.
    [12]. Neuenschwander S, Hoerstrup SP. Heart valve tissue engineering. Transplant Immunology,2004,12(3-4):359-365.
    [13]. Fuchs JR., Nasseri BA, Vacanti JP. Tissue engineering:a 21st century solution to surgical reconstruction. Ann Thorac Surg,2001,72(2):577-591.
    [14]. Gu CH, Wei XF, Wang YY, et al. No infection with porcine endogenous retrovirus in recipients of acellular porcine aortic valves:a two-year study. Xenotransplantation, 2008,15(2):121-128.
    [15]. Wilczek P. Heart valve bioprothesis; effect of different acellularizations methods on the biomechanical and morphological properties of porcine aortic and pulmonary valve. Bulletin of the Polish Academy of Science-Technical Sciences,2010,58(2): 337-342.
    [16]. Schenke-Layland K, Vasilevski O, Opitz F, et al. Impact of decellularization of xenogeneic tissue on extracellular matrix integrity for tissue engineering of heart valves. Journal of Structural Biology,2003,143(3):201-208.
    [17].董念国,叶晓峰,孙宗全,等。脱细胞天然支架生物力学性能变化及预处理改善组织相容性的实验研究.中华外科杂志,2007,45(16):1128-1131.
    [18]. Liao J, Joyce EM, Sacks MS. Effects of decellularization on the mechanical and structural properties of the porcine aortic valve leaflet. Biomaterials,2008,29(8): 1065-1074.
    [19]. Raeber GP, Lutolf MP, Hubbell JA. Molecularly engineered peg hydrogels:a novel model system for proteolytically mediated cell migration. Biophysical J,2005,89: 1374-88.
    [20]. Porter AM, Klinge CM, Gobin AS. Biomimetic Hydrogels with VEGF Induce Angiogenic Processes in Both hUVEC and hMEC. Biomacromolecules,2011,12(1): 242-246.
    [21]. Harmand MF. In vitro study of biodegradation of Co-Cr alloy using a human cell mode. J Biomater Sci Polym Ed,1995,6(9):809.
    [22].奚延裴.组织工程医疗产品的安全性评价.现代康复,2001,5(6):13-15.
    [23]. ISO 10993-5:1999, Biological evaluation of medical devices-Part 5:Tests for in vitro cytotoxicity.
    [1]. Bader A, Schilling T, Teebken OE, et al. Tissue engineering of heart valves-human endothelial cell seeding of detergent acellularized porcine valves. Eur J Cardiothorac surg,1999,14:279-284.
    [2]. Booth C, Korossis SA, Wilcox HE, et al, Tissue engineering of cardiac valve prostheses I:development and histological characterization of an acellular porcine scaffold. J Heart Valve Dis,2002,11(4):457-462.
    [3]. Schenke-Layland K, Vasilevski O, Opitz F, et al. Impact of decellularization of xenogeneic tissue on extracellular matrix integrity for tissue engineering of heart valves. J Struct Biol,2003,143(3):201-208.
    [4].董念国,叶晓峰,孙宗全,等。脱细胞天然支架生物力学性能变化及预处理改善组织相容性的实验研究.中华外科杂志,2007,45(16):1128-1131.
    [5]. Liao J, Joyce E M, Sacks M S. Effects of decellularization on the mechanical and structural properties of the porcine aortic valve leaflet. Biomaterials,2008,29(8): 1065-1074.
    [6]. Flanagan TC, Pandit A. Living artificial heart valve alternatives. Euro Cell Mater, 2003,6:28-45.
    [7]. Hersel U, Dahmen C, Kessler H. RGD modified polymers:biomaterials for stimulated cell-adhesion and beyond. Biomaterials,2003,24(24):4385-4415.
    [8]. Shi JW, Dong NG, Sun ZQ. Immobilization of Decellularized Valve Scaffolds with Arg-Gly-Asp-containing Peptide to Promote Myofibroblast Adhesion. J Huazhong Univ Sci Technol,2009,29(4):503-507.
    [9]. Dong XC, Wei XF, Yi W, et al. RGD-modified acellular bovine pericardium as a bioprosthetic scaffold for tissue engineering. J Mater Sci Mater Med,2009,20: 2327-2336.
    [10]. Shachar M, Tsur-Gang O, Dvir T, et al. The effect of immobilized RGD peptide in alginate scaffolds on cardiac tissue engineering. Acta Biomateriala,2011,7(1): 152-162.
    [11].陈心,罗素兰,张本,等.多肽固相合成的研究进展.生物技术,2006,16(1):81-82
    [12]. Fields GB, Noble RL. Solid phase peptide synthesis utilizing 9-fluorenylmetho-xycarbonyl amino acids. Int J Peptide Protein Res,1990,35(3):161-214.
    [13]. Cudic P, Stawikowski M. Peptidomimetics:fmoc solid-phase pseudopeptide synthesis. Methods Mol Biol,2008,494:223-246.
    [14]. Liu Z, Hu BH, Messersmith PB. Convenient Synthesis of Acetonide Protected 3, 4-Dihydroxyphenylalanine (DOPA) for Fmoc Solid-Phase Peptide Synthesis. Tetrahedron Lett,2008,49(38):5519-5521.
    [15]. Mullen DG, Kyro K, Hauser M, et al. Synthesis of a-factor peptide from Saccharomyces cerevisiae and photoactive analogues via Fmoc solid phase methodology.2011,19(1):490-497.
    [16]. Mather BD, Viswanathan K, Miller KM, et al. Michael addition reactions in macromolecular design for emerging technologies. Progress in Polymer Science,2006, 31(5):487-531.
    [17]. Zhu J, Tang C, Kottke-Marchant K, et al. Design and synthesis of biomimetic hydrogel scaffolds with controlled organization of cyclic RGD peptides. Bioconjug Chem,2009,20(2):333-339.
    [18]. Leung DW, Cachines G, Kuang WJ, et al. Vascular endothelial growth factor issecreted angiogenic mitogen. Science,1989,246:1306.
    [19]. Neufeld G, Cohen T, Stela G, et al. Vascular endothelial growth factor (VEGF) and its receptors. FASEB J,1999,13:9-22.
    [20]. Lutolf MP, Tirelli N, Cerritelli S, et al. Systematic modulation of Michael-type reactivity of thiols through the use of charged amino acids. Bioconjuate Chem,2001, 12(6):1051-1056.
    [21]. Zisch AH, Lutolf MP, Ehrbar M, et al. Cell-demanded release of VEGF from synthetic, biointeractive cell-ingrowth matrices for vascularized tissue growth. FASEB J,2003,17:2260-2262.
    [22]. Hu Y, Winn SR, Krajbich I, et al. Porous polymer scaffolds surface-modified with arginine-glycine-aspartic acid enhance bone cell attachment and differentiation in vitro. J Biomed Mater Res,2003,64A:583-590.
    [23]. Guo LM, Zeng XF, Ma RD, et al. Surface modification of RGD peptides onto acellularized porcine aortic valve to promote cell adhesion. Sichuan Da Xue Xue Bao Yi Xue Ban,2010,41(6):1008-11,1054.
    [24]. Zhang Z, Lai YX, Yu L, et al. Effects of immobilizing sites of RGD peptides in amphiphilic block copolymers on efficacy of cell adhesion. Biomaterials,2010, 31(31):7873-7882.
    [25]. Chollet C, Lazare S, Guillemot F, et al. Impact of RGD micro-patterns on cell adhesion. Colloids and Surfaces B:Biointerfaces,2010,75(1):107-114.
    [26]. Zisch AH, Lutolf MP, Ehrbar M, et al. Cell-demanded release of VEGF from synthetic, biointeractive cell ingrowth matrices for vascularized tissue growth. The FASEB Journal,2003,17:2260-2262.
    [27]. Ferrara N, Houck K, Jakeman L, et al. Molecular and biological properties of the vascular endothelial growth factor family of proteins. Endocrine Reviews,1992,13 (1):18-32.
    [28]. Porter AM, Klinge CM, Gobin AS. Biomimetic Hydrogels with VEGF Induce Angiogenic Processes in Both hUVEC and hMEC. Biomacromolecules,2011,12(1): 242-246.
    [1]. Sacks MS. Biomechanics of engineered heart valve tissues. Conf Proc IEEE Eng Med Biol Soc,2006,1:853-854.
    [2]. Asahara T, Murohara T, Sullivan A, et al. Isolation of putative progenitor endothelial cells for angiogenesis. Science,1997,275(5302):964-967.
    [3]. Park SJ, Baek SH, Oh MK, et al. Enhancement of angiogenic and vasculogenic potential of endothelial progenitor cells by haptoglobin. FEBS Letters,2009,583: 3235-3240.
    [4]. Baudin B, Bruneel A, Bosselut N, et al. A protocol for isolation and culture of human umbilical vein endothelial cells. Nat Protoc,2007,2 (3):481-485.
    [5]. Sodian R, Hoerstrup SP, Sperling JS, et al. Tissue engineering of heart valves:in vitro experiences. Ann Thorac Surg,2000,70(1):140-144.
    [6]. Cebotari S, Mertsching H, Kallenbach K, et al. Construction of autologous human heart valves based on an acellular allograft matrix. Circulation,2002,106(3):63-68.
    [7]. Perry TE, Kaushal S, Sutherland FWH, et al. Bone marrow as a cell source for tissue engineering heart valve. Ann Thorac Surg,2003,75(3):761-767.
    [8]. Neuenschwander S, Hoerstrup SP. Heart valve tissue engineering. Transplant Immunology,2004,12(3-4):359-365.
    [9]. Asahara T, Murohara T, Sullivan A, et al. Isolation of putative progenitor endothelial cells for angiogenesis. Science,1997,275(5302):964-967.
    [10]. Kaushal S, Amiel GE, Guleserian KJ, et al. Functional small-diameter neovessels created using endothelial progenitor cells expanded ex vivo. Nat Med,2001,7(9): 1035-1040.
    [11]. Peichev M, Naiyer AJ, Pereira D, et al. Expression of VEGFR-2 and AC133 by circulating human CD34(+) cells identifies a population of functional endothelial precursors. Blood,2000,95(3):952-958.
    [12]. Rocha V, Wagner JE Jr, Sobocinski KA, et al. Graft-versus-host disease in children who have received a cord-blood or bone marrow transplant from an HLA-identical sibling. N Engl J Med,2000,342(25):1846-1854.
    [13]. Steve K, Zhu YQ, Yang GY. Preparation and analysis of endothelial progenitor cells associated with angiogenesis. Protocols for Adult Stem Cells,2010,139-148.
    [14]. Timmermans F, Plum J, Yoder MC, et al. Endothelial progenitor cells:identity defined? J Cell Mol Med,2009,13(1):87-102.
    [15]. Gehling UM, Ergun S, Schumacher U, et al. In vitro differentiation of endothelial cells from AC133-positive progenitor cells. Blood,2000,95(10):3106-3112.
    [16]. Song E, Lu CW, Fang LJ, et al. Culture and identification of endothelial progenitor cells from human umbilical cord blood. International Journal of Ophthalmology, 2010,3(1):49-53.
    [17]. Motwani MS, Rafiei Y, Tzifa A, et al. In situ endothelialization of intravascular stents from progenitor stem cells coated with nanocomposite and functionalized biomolecules. Biotechnology and Applied Biochemistry,2011,57:2-13.
    [18]. Ong LL, Li W, Oldigs JK, et al. Hypoxic/normoxic preconditioning increases endothelial differentiation potential of human bone marrow CD 133+ cells. Tissue Eng Part C Methods,2010,16(5):1069-1081.
    [19]. Sales VL, Engelmayr GC Jr, Mettler BA, et al. Transforming growth factor-betal modulates extracellular matrix production, proliferation, and apoptosis of endothelial progenitor cells in tissue-engineering scaffolds. Circulation,2006,114(1 Suppl):193-199.
    [1]. 司徒镇强,吴军正.细胞培养.西安:世界图书出版社,2004.259-261.
    [2]. 刘刚,胡蕴玉,颜永年,等.Ⅰ型胶原修饰多孔材料聚乙醇酸乳酸共聚物对兔骨髓间充质干细胞黏附和增殖及成骨细胞基因的表达影响.中华医学杂志,2003,83(7):580-583.
    [3]. Fang NT, Xie SZ, Wang SM, et al. Construction of tissue-engineered heart valves by using decellularized scaffolds and endothelial progenitor cells. Chin Med J,2007, 120(8):696-702.
    [4]. Kasimir MT, Rieder E, Seebacher G, et al. Decellularization does not eliminate thrombogenicity and inflammatory stimulation in tissue-engineered porcine heart valves. J Heart Valve Dis,2006,15(2):278-86.
    [5]. Kasimir MT, Weigel G, Sharma J, et al. The decellularized porcine heart valve matrix in tissue engineering:platelet adhesion and activation. Thromb Haemost, 2005,94:562-567.
    [6]. Dohmen PM, Ozaki S, Nitsch R, et al. A tissue engineered heart valve implanted in a juvenile sheep model. Med Sci Monit,2003,9(4):BR97-BR104.
    [7]. Dohmen PM, Lembcke A, Hotz H, et al. Ross operation with a tissue-engineered heart valve. Ann Thorac Surg,2002,74:1438-1442.
    [8]. Breuer CK, Mettler BA, Anthony T, et al. Application of tissue-engineering principles toward the development of a semilunar heart valve substitute. Tissue Eng, 2004,10(11-12):1725-1736.
    [9]. Kasimir MT, Weigel G, Sharma J, et al. The decellularized porcine heart valve matrix in tissue engineering:platelet adhesion and activation. Thromb Haemost, 2005,94:562-567.
    [10]. Hersel U, Dahmen C, Kessler H. RGD modified polymers:biomaterials for stimulated cell adhesion and beyond. Biomaterials,2003,24(24):4385-4415.
    [11]. Wang H, Ma L, Yang SH, et al. Effect of RGD-modified Silk Material on the Adhesion and Proliferation of Bone Marrow-derived Mesenchymal Stem Cells. J Huazhong Univ Sci Technol,2009,29(1):80-83.
    [12]. Breuer CK, Mettler BA, Anthony T, et al. Application of tissue-engineering principles toward the development of a semilunar heart valve substitute. Tissue Eng, 2004,10(11-12):1725-1736.
    [13]. Tran NQ, Joung YK, Lih E, et al. RGD-conjugated In Situ forming hydrogels as cell-adhesive injectable scaffolds. Macromolecular Research,2011,19(3):300-306.
    [14]. Shi JW, Dong NG, Sun ZQ. Immobilization of Decellularized Valve Scaffolds with Arg-Gly-Asp-containing Peptide to Promote Myofibroblast Adhesion.J Huazhong Univ Sci Technol,2009,29(4):503-507.
    [15]. Lodish H, Berk A, Zipursky SL, et al. Molecular Cell Biology.3rd Ed. Freeman WH. New York:1995.1247-1294.
    [16]. Dong XC, Wei XF, Yi W, et al. RGD-modified acellular bovine pericardium as a bioprosthetic scaffold for tissue engineering. J Mater Sci:Mater Med,2009,20: 2327-2336.
    [17]. Li CW, Zheng QX, Guo XD, et al. Combined Use of RGD-peptide Modified PLGA and TGF-β1 Gene Transfected MSCs to Improve Cell Biobehaviors in vitro. J Huazhong Univ Sci Technol,2009,29(5):592-598.
    [18]. Neufeld G, Cohen T, Stela G, et al. Vascular endothelial growth factor (VEGF) and its receptors. FASEB J,1999,13:9-22.
    [19]. Knetsch MLW, Koole LH. VEGF-E enhances endothelialization and inhibits thrombus formation on polymeric surfaces. Journal of Biomedical Materials Research,2010,93A(1):77-85.
    [20]. Muller S, Koenig G, Charpiot A, et al, VEGF-Functionalized Polyelectrolyte Multilayers as Proangiogenic Prosthetic Coatings. Advance Functional Materials, 2008,18(12):1767-1775.
    [21]. Shen YH, Shoichet MS, Radisic M. Vascular endothelial growth factor immobilized in collagen scaffold promotes penetration and proliferation of endothelial cells. Acta Biomaterialia,2008,4(3):477-489.
    [22]. Poter AM, Klinge CM, Gobin AS. Biomimetic Hydrogels with VEGF Induce Angiogenic Processes in Both hUVEC and hMEC. Biomacromolecules,2011,12(1): 242-246.
    [23]. Zisch AH, Lutolf MP, Ehrbar M, et al. Cell-demanded release of VEGF from synthetic, biointeractive cell-ingrowth matrices for vascularized tissue growth. FASEB J,2003,17:2260-2262.
    [24]. Poh CK, Shi ZL. Lim TY, et al. The effect of VEGF functionalization of titanium on endothelial cells in vitro. Biomaterials,2010,31(7):1578-1585.
    [25]. Leach LK, Kaigle D, Wang Z, et al. Coating of VEGF-releasing scaffolds with bioactive glass for angiogenesis and bone regeneration. Biomaterials,2006,27(17): 3249-3255.
    [26]. Shinkai A, Ito M, Anazawa H, et al. Mapping of the sites involved in ligand association and dissociation at the extracellular domain of the kinase insert domaincontaining receptor for vascular endothelial growth factor. J Biol Chem, 1998,273:31283-31288.
    [27]. Shibuya M. Vascular endothelial growth factor receptor-2:its unique signaling and specifc ligand, VEGF-E. Cancer Sci,2003,94(9):751-756.
    [28]. Holmes K, Roberts OL, Thomas AM, et al. Vascular endothelial growth factor receptor-2:structure, function, intracellular signaling and therapeutic inhibition. Cell Signal,2007,19(10):2003-2012.
    [29]. Raeber GP, Lutolf MP, Hubbell JA. Molecularly engineered peg hydrogels:a novel model system for proteolytically mediated cell migration. Biophysical J,2005, 89(2):1374-1388.
    [30]. Porter AM, Klinge CM, Gobin AS. Biomimetic Hydrogels with VEGF Induce Angiogenic Processes in Both hUVEC and hMEC. Biomacromolecules,2011,12: 242-246.
    [31].邓诚,董念国,史嘉玮,等.聚乙二醇载药缓释微球复合去细胞瓣制备组织工程心脏瓣膜支架.临床心血管病杂志,2010,26(1):68-71.
    [32]. Ouyang H, Zhang JB, Liu Q, et al. Research on application of modified polyethylene glycol hydrogels in the construction of tissue engineered heart valve. Zhonghua Wai Ke Za Zhi,2008,46(22):1723-6.
    [33]. Zisch AH, Lutolf MP, Ehrbar M, et al. Cell-demanded release of VEGF from synthetic, biointeractive cell-ingrowth matrices for vascularized tissue growth. FASEB J,2003,17:2260-2262.
    [34]. Raeber GP, Lutolf MP, Hubbell JA. Molecularly Engineered PEG Hydrogels:A Novel Model System for Proteolytically Mediated Cell Migration. Biophys J,2005, 89(2):1374-1388.
    [35]. Lutolf MP, Tirclli N, Cerritelli S, et al. Systematic modulation of Michael-type reactivity of thiols through the use of charged amino acids. Bioconjuate Chem.2001, 12(6):1051-1056.
    [36]. Lutolf MP, Hubbell JA. Synthetic biomaterials as instructive extracellular microenvironments for morphogenesis in tissue engineering. Nat Biotechnol,2005, 23(1):47-55.
    [37]. Xing FY, Jiang Y, Liu J, et al. Role of API Element in the Activation of Human eNOS Promoter by Lysophosphatidycholine, Journal of Cellular Biochemistry,2006, 98(2):872-884.
    [38]. Wen HK, Jun TH, Zheng FB, et al. p38 MAPK-dependent eNOS upregulation is critical for 17β-estradiolmediated cardioprotection following trauma-hemorrhage. Am J Physiol Heart Circ Physiol,2008,294(6):2627-2636.
    [39]. Naoki S, Salvatore S, Hyung-Hwan K, et al. Regulation of enduthelial nitric oxide synthase and postnatal angiogenesis by Racl. Circ Re s,2008,4:360-368.
    [40]. Yang Z, Tao J, Wang JM, et al. Shear stress contributes to t-PA mRNA expression in human endothelial progenitor cells and nonthrombogenic potential of small diameter artificial vessels. Biochem Biophys Res Commun,2006,342(2):577-584.
    [41]. Fang NT, Xie SZ, Wang SM, et al. Construction of tissue-engineered heart valves by using decellularized scaffolds and endothelial progenitor cells. Chin Med J,2007, 120(8):696-702.
    [42]. Suh W, Kim KL, Choi JH, et al. C-reactive protein impairs angiogenic functions and decreases the secretion of arteriogenic chemo-cytokines in human endothelial progenitor cells. Biochem Biophys Res Commun,2004,321(1):65-71.
    [1]. Jordan SW, Chaikof EL. Novel thromboresistant materials. J Vasc Surg,2007,45: A104-A115.
    [2]. Kidane AG, Salacinski H, Tiwari A, et al. Anticoagulant and antiplatelet agents:their clinical and device application(s) together with usages to engineer surfaces. Biomacromolecules,2004,5:798-813.
    [3]. Kasimir MT, Rieder E, Seebacher G, et al. Decellularization does not eliminate thrombogenicity and inflammatory stimulation in tissue-engineered porcine heart valves. J Heart Valve Dis,2006,15(2):278-86.
    [4]. Kasimir MT, Wei gel G, Sharma J, et al. The decellularized porcine heart valve matrix in tissue engineering:platelet adhesion and activation. Thromb Haemost,2005, 94:562-567.
    [5]. Dohmen PM, Ozaki S, Nitsch R, et al. A tissue engineered heart valve implanted in a juvenile sheep model. Med Sci Monit,2003,9:BR97-BR104.
    [6]. Dohmen PM, Lembcke A, Hotz H, et al. Ross operation with a tissue-engineered heart valve. Ann Thorac Surg,2002,74:1438-1442.
    [7]. Schopka S, Schmid T, Schmid C, et al. Current strategies in cardiovascular biomaterial functionalization. Materials,2010,3:638-655.
    [8]. Breuer CK, Mettler BA, Anthony T, et al. Application of tissue-engineering principles toward the development of a semilunar heart valve substitute. Tissue Eng,2004,10: 1725-1736.
    [9]. Bordenave L, Fernandez P, Remy-Zolghadri M, et al. In vitro endothelialized ePTFE prostheses:Clinical update 20 years after the first realization. Clin Hemorheol Microcirc,2005,33:227-234.
    [10]. Kleinman HK, Philp D, Hoffman MP. Role of the extracellular matrix in morphogenesis. Current Opinion in Biotechnology,2003,14(5):526-532.
    [11]. Shi JW, Dong NG, Sun ZQ. Immobilization of Decellularized Valve Scaffolds with Arg-Gly-Asp-containing Peptide to Promote Myofibroblast Adhesion.J Huazhong Univ Sci Technol,2009,29(4):503-507.
    [12]. Lutolf MP, Hubbell JA. Synthetic biomaterials as instructive extracellular microenvironments for morphogenesis in tissue engineering. Nature Biotechnology, 2005,23:47-55.
    [13]. Dong XC, Wei XF, Yi W, et al. RGD-modified acellular bovine pericardium as a bioprosthetic scaffold for tissue engineering. J Mater Sci:Mater Med,2009,20: 2327-2336.
    [14].Hong H, Dong GN, Shi WJ,et al. Fabrication of biomatrix/polymer hybrid scaffold for heart valve tissue engineering in vitro. ASAIO J,2008 Nov-Dec,54(6):627-632.
    [15].邓诚,董念国,史嘉玮,等.聚乙二醇载药缓释微球复合去细胞瓣制备组织工程心脏瓣膜支架.临床心血管病杂志,2010,26(1):68-71.
    [16].董念国,叶晓峰,孙宗全,等。脱细胞天然支架生物力学性能变化及预处理改善组织相容性的实验研究.中华外科杂志,2007,45(16):1128-1131。
    [17]. Hersel U, Dahmen C, Kessler H. RGD modified polymers:biomaterials for stimulated cell adhesion and beyond. Biomaterials,2003,24:4385-4415.
    [18]. Li CW, Zheng QX, Guo XD, et al. Combined Use of RGD-peptide Modified PLGA and TGF-β1 Gene Transfected MSCs to Improve Cell Biobehaviors in vitro. J Huazhong Univ Sci Technol,2009,29(5):592-598.
    [19]. Wang H, Ma L, Yang SH, et al. Effect of RGD-modified Silk Material on the Adhesion and Proliferation of Bone Marrow-derived Mesenchymal Stem Cells. J Huazhong Univ Sci Technol,2009,29(1):80-83.
    [20]. Alobaid N, Salacinski HJ, Sales KM, et al. Nanocomposite Containing Bioactive Peptides Promote Endothelialisation by Circulating Progenitor Cells:An In vitro Evaluation. European Journal of Vascular & Endovascular Surgery,2006,32(1): 76-83.
    [21]. Tran NQ, Joung YK, Lih E, et al. RGD-conjugated In Situ forming hydrogels as cell-adhesive injectable scaffolds. Macromolecular Research,2011,19(3):300-306.
    [22]. Stegemann JP, Kaszuba SN, Rowe SL. Review:Advances in Vascular Tissue Engineering Using Protein-Based Biomaterials. Tissue Eng,2007,13(11): 2601-2613.
    [23]. Heilshorn SC, DiZio KA, Welsh ER, et al. Endothelial cell adhesion to the fibronectin CS5 domain in artificial extracellular matrix proteins. Biomaterials,2003,24(23): 4245-4252.
    [24]. Shi JW, Dong NG, Sun ZQ. Immobilization of Decellularized Valve Scaffolds with Arg-Gly-Asp-containing Peptide to Promote Myofibroblast Adhesion.J Huazhong Univ Sci Technol,2009,29(4):503-507.
    [25]. Dong XC, Wei XF, Yi W, et al. RGD-modified acellular bovine pericardium as a bioprosthetic scaffold for tissue engineering. J Mater Sci:Mater Med,2009,20: 2327-2336.
    [26]. Li CW, Zheng QX, Guo XD, et al. Combined Use of RGD-peptide Modified PLGA and TGF-β1 Gene Transfected MSCs to Improve Cell Biobehaviors in vitro. J Huazhong Univ Sci Technol,2009,29(5):592-598.
    [27]. Wang H, Ma L, Yang SH, et al. Effect of RGD-modified Silk Material on the Adhesion and Proliferation of Bone Marrow-derived Mesenchymal Stem Cells. J Huazhong Univ Sci Technol,2009,29(1):80-83.
    [28]. Hersel U, Dahmen C, Kessler H. RGD modified polymers:Biomaterials for stimulated cell adhesion and beyond. Biomaterials,2003,24:4385-4415.
    [29]. Tran NQ, Joung YK, Lih E, et al. RGD-conjugated In Situ forming hydrogels as cell-adhesive injectable scaffolds. Macromolecular Research,2011,19(3):300-306.
    [30]. Hatakeyama H, Kikuchi A, Yamato M, et al. Bio-functionalized thermoresponsive interfaces facilitating cell adhesion and proliferation. Biomaterials,2006,27(29): 5069-5078.
    [31]. Leung DW, Cachines G, Kuang WJ, et al. Vascular endothelial growth factor issecreted angiogenic mitogen[J]. Science,1989,246:1306.
    [32]. Neufeld G, Cohen T, Stela G, et al. Vascular endothelial growth factor(VEGF) and its receptors. FASEB J,1999,13:9-22.
    [33]. Holmes K, Roberts OL, Thomas AM, et al. Vascular endothelial growth factor receptor-2:structure, function, intracellular signaling and therapeutic inhibition. Cell Signal,2007,19(10):2003-2012.
    [34]. Knetsch MLW, Koole LH. VEGF-E enhances endothelialization and inhibits thrombus formation on polymeric surfaces. Journal of Biomedical Materials Research, 2010,93A(1):77-85.
    [35]. Muller S, Koenig G, Charpiot A, et al, VEGF-Functionalized Polyelectrolyte Multilayers as Proangiogenic Prosthetic Coatings. Advance Functional Materials, 2008,18(12):1767-1775.
    [36]. Shen YH, Shoichet MS, Radisic M. Vascular endothelial growth factor immobilized in collagen sca□old promotes penetration and proliferation of endothelial cells. Acta Biomaterialia,2008,4:477-489.
    [37]. Poter AM, Klinge CM, Gobin AS. Biomimetic Hydrogels with VEGF induce angiogenic processes in both hUVEC and hMEC. Biomacromolecules,2011,12: 242-246.
    [38]. Zisch AH, Lutolf MP, Ehrbar M, et al. Cell-demanded release of VEGF from synthetic, biointeractive cell-ingrowth matrices for vascularized tissue growth. FASEB J,2003,17:2260-2262.
    [39]. Poh CK, Shi ZL. Lim TY, et al. The effect of VEGF functionalization of titanium on endothelial cells in vitro. Biomaterials,2010,31(7):1578-1585.
    [40]. Leach LK, Kaigle D, Wang Z, et al. Coating of VEGF-releasing scaffolds with bioactive glass for angiogenesis and bone regeneration. Biomaterials,2006,27(17): 3249-3255.
    [41]. Walpoth BH, Zammaretti P, Cikirikcioglu M, et al. Enhanced intimal thickening of expanded polytetrafluoroethylene grafts coated with fibrin or fibrin-releasing vascular endothelial growth factor in the pig carotid artery interposition model. J Thorac CardioVasc Surg,2007,133(5):1163-1170.
    [42]. Bunting S, Moncada S, Vane JR. Antithrombotic properties of vascular endothelium. Lancet,1977,2:1075-1076.
    [43]. De Mel A, Jell G, Stevens MM, et al. Biofunctionalization of biomaterials for accelerated in situ endothelialization:a review. Biomacromolecules,2008,9: 2969-2679.
    [44]. Melero-Martin JM, Khan ZA, Picard A, et al. In vivo vasculogenic potential of human blood-derived endothelial progenitor cells. Blood,2007,109:4761-4768.
    [45]. Rothmans JI, Heyligers JMM, Stroes ESG, et al. Endothelial progenitor cell-seeded grafts:Rash and risky. Can J Cardiol,2006,22:929-932.
    [46]. Dohmen PM, Lembcke A, Hotz H, et al. Ross operation with a tissue-engineered heart valve. Ann Thorac Surg,2002,74:1438-1442.
    [47]. Schmidt D, Breymann C, Weber A, et al. Umbilical cord blood derived endothelial progenitor cells for tissue engineering of vascular grafts. Ann Thorac Surg,2004,78: 2094-2098.
    [48]. Schmidt D, Mol A, Neuenschwander S, et al. Living patches engineered from human umbilical cord derived fibroblasts and endothelial progenitor cells. Eur J Cardiothorac Surg,2005,27:795-800.
    [49]. Kaushal S, Amiel GE, Guleserian KJ, et al. Functional small diameter neovessels created using endothelial progenitor cells expanded in vivo. Nat Med,2001,7: 1035-1040.
    [50]. Avci-Adali M, Paul A, Ziemer G, et al. New strategies for in vivo tissue engineering by mimicry of homing factors for self-endothelialization of blood contacting materials. Biomaterials,2008,29:3936-3945.
    [51]. Rodenberg EJ, Pavalko FM. Peptides Derived from Fibronectin Type III Connecting Segments Promote Endothelial Cell Adhesion but Not Platelet Adhesion:Implications in Tissue-Engineered Vascular Grafts. Tissue Eng,2007,13(11):2653-2666.
    [52]. Moncada S, Palmer RM, Higgs EA. The discovery of nitric oxide as the endogenous nitrovasodilator. Lancet,1987,2:1057-1058.
    [53]. Lev EI, Estrov Z, Aboulfatova K, et al. Potential role of activated platelets in homing of human endothelial progenitor cells to subendothelial matrix. Thromb Haemost, 2006,96:498-504.

© 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