用户名: 密码: 验证码:
皮肤组织工程新型材料的合成及表面改性的生物学基础研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
PLGA是一种无毒可降解的高分子有机聚合物,其在机体内代谢首先通过聚酯水解为乳酸和羟基乙酸,随后被完全分解成为二氧化碳和水排出体外,生物相容性好。PVA水凝作为人工敷料的优点是含水量丰富,不但可以保持创面湿润,还具有吸水性可以吸收组织渗出液,同时其内部大量贯通的孔隙有利于营养物质和代谢废物的运输,且又镇痛作用,因而广泛应用与组织工程领域。本研究综合了两者的优势,并采用物理交联的方式将二者复合,构建一种类似皮肤结构的双层人工支架,使之更适合于临床骨皮肤损伤、修复的需要。
     明胶提取自动物的骨头或结缔组织,无毒无害,被广泛用于食品、制药及组织工程领域。在皮肤组织工程领域研究中,明胶除了具有促进细胞粘附和生长的作用,还可以作为一种底物基质与细胞外基质产生竞争作用,防止伤口部位的蛋白酶消化细胞外基质。明胶分子结构上含有大量的羟基及少量的羧基和氨基,具有极强的亲水性。因此,明胶分子中的氨基可以与光活性叠氮基团发生缩合反应,形成酯类。本研究为进一步提高人工皮肤支架材料的生物活性,以组织工程生物材料为基底,探讨光固定表面改性的对其生物相容性的影响。
     结果显示:本研究利用光活性叠氮基团与明胶分子的羧基发生了缩合反应,形成具有光活性的新的化合物—Az-G光活性明胶;通过在材料表面的固定效果和细胞学评价,证明Az-G光活性明胶可以稳定的固定在生物材料表面,不会脱落,且细胞相容性好,有利于细胞粘附、生长和增殖;本研究首次将静电纺丝膜和PVA水凝胶通过物理冷冻交联的方法结合,其机型性能和保水性远优于单纯的PLGA电纺丝膜或PVA水凝胶;采用模具法制备的“多孔”PVA水凝胶有利于细胞的粘附和生长。
Continuous self-renewal of normal skin epidermis, dermis and subcutaneoussubsidiary organ damage by external impact, through its own physiologicalregeneration and repair of skin tissue, namely compensatory regeneration. However,for large area burns, soft tissue trauma, congenital nevus and skin necrosis diseasewill lead to full-thickness skin defects, skin normal physiological repair systems facesignificant challenges, has been unable to meet the need for tissue regeneration, skintransplant. Supply patients with autologous the defect covered most traditional skingraft technique is a single-layer skin transplantation, which is currently the clinicalgold standard of treatment. This type of single-layer contains all skin epidermal tissuestructures, but only a portion of the leather structure, often resulting in the formationof scar. Therefore, the development of a full-thickness skin of normal anatomy andfunction of the skin analogues is the solution to donor shortage of skin and scarformation is the most effective way.
     PLGA is a non-toxic biodegradable high molecular weight organic polymer, itsmetabolism in the body through the first polyester hydrolysis to lactic acid andglycolic acid, and subsequently excreted completely decomposed into carbon dioxideand water, good biocompatibility, has passed U.S. FDA, was formally included aspharmaceutical excipients into the United States Pharmacopoeia, widely used inpharmaceutical, medical engineering materials and modern industrial fields. Is oftenused in tissue engineering and regenerative medicine the electrospinning film madetherefrom, is considered to be a two-dimensional bracket having goodbiocompatibility. PVA hydrogel as the advantages of artificial dressing is rich inwater content, can not only keep the wound moist, absorbent can absorb tissueexudate, at the same time, the interior of a large number of through pores in favor of nutrients and metabolic waste transport, and because the town pain effect, so widelyused in the field of tissue engineering. This study combines the advantages of both,and physical cross-linking of the two composite a double stent to build a skin-likestructure, to make it more suitable for clinical bone skin damage repair needs.
     Gelatin extracted from animal bone or connective tissue, non-toxic, is widelyused in food, pharmaceutical and tissue engineering. In skin tissue engineeringresearch in the field, the gelatin in addition to having a role in promoting cell adhesionand growth, and can also be used as a substrate matrix extracellular matrix resulting incompetitive effect, prevent the wound site of the protease to digest the extracellularmatrix. Past, gelatin is mainly based on physical mixing applications in tissueengineering, thereby enhancing the biocompatibility of the scaffold. Although thismethod is simple, can improve the biocompatibility of the material to a certain extent,but only through the coating, physical adsorption, or a method of mixing a materialsurface modified poor stability, easy to fall off. Currently, the surface modificationmethod of tissue engineering materials including surface finishing, physicaladsorption, and the surface of the plasma polymerization, photochemical fixed.
     Compared with the other biological material surface modification method, lightfixed technology has prominent features:①both ideal surface interface performance,but also the nature of the material itself does not affect;②the operation is simple,rapid response, and low cost;③versatility, suitable for almost all biological polymermaterials;④can be any target molecule into derivatives with photoactive groups, andphotolysis conditions are very similar;⑤light fixed a variety of biological moleculeshaving good stability, and its biological activity is almost unaffected; The⑥apply asurface modification of the complex material in a variety of shapes, such as the use ofquartz optical fiber guide ultraviolet, above and according to the need for any part ofthe surface of the material by size or regional processing to obtain medical polymermaterials to meet different clinical requirements;⑦can greatly reduce the purpose ofcoating reagents disorderly medical polymer materials surface cross-linking, the targetmolecules in the surface of the material in a highly ordered state, to avoid the use of other methods, the activity multiplied loss of the physical and biological properties ofthe coating and the surface of the biological material and other surface propertiesgreatly reduced defect.
     The gelatin is collagen hydrolyzate, and its main by the same composition andmolecular weight distribution of a wide amino acids, and generally has a molecularweight in the tens of thousands of ten thousands. Gelatin on the molecular structurecontains a large number of hydroxyl groups and a small amount of carboxyl groupand an amino group, with a strong hydrophilic. Thus, the amino group in the gelatinmolecule can occur with the azide group of the photoactive condensation reaction toform esters.
     Accordingly, the present study was to further enhance the biological activity ofthe scaffold material of the artificial skin substrate to investigate the impact of lightfixed to its surface-modified biocompatible biological material for tissue engineering.
     一、Preparation and characterization of a photoactive gelatin
     1. The Synthesis and Characterization of Az-G photoactive gelatin
     Through the activation of carboxyl group, the condensation reaction, dialysis andfreeze-dried after a series of processes, the photoactive gelatin prepared Az-G, thephotoactive chemical composition was visually observed as a white flocculent solid,soft and sticky.
     Respectively, using ultraviolet spectroscopy, infrared spectroscopy and nuclearmagnetic resonance of three methods, the characterization of the molecular structureof the photoactive gelatin Synthesis of Az-G: a multifunctional microplate reader,measured within the range of230-800nm gelatin, AZ-G an ultraviolet absorptionspectrum of the photoactive gelatin and4-azido benzoic acid results in the UVabsorption spectra at268nm wavelength,4-azido acid azide group has characteristicpeaks of pure gelatin is not displayed any absorption peak, and Az-G photoactivegelatin characteristic peak appears at270nm; Fourier transform infrared spectroscopycompare the structural differences of the Az-G photoactive gelatin and the gelatinalone, the synthetic compound of the gelatin in the2100nm at a wavelength of a downward peaks, the peaks belonging to the characteristic peaks of the azide does notappear pure gelatin IR spectra of the peaks; NMR results show, the the gelatincompounds obtained by the condensation reaction appears on the position of theδ7.8917,7.7263the characteristic peaks in the benzene ring peak, while the the puregelatin map not the peaks.
     2. Az-G photoactive gelatin preferred crosslinking reaction conditions
     Reference Synthesis of Az-G the photoactive gelatin having a photoactive bytheir molecular structure and therefore using ultraviolet spectrometer aqueous gelatinsolution before and after the ultraviolet radiation photoactive full wavelength scan,through the observation of the type of radiation intensity, the type of radiation time ofthe optical activity collagen, thus worked best photoactive gelatin crosslinkingreaction conditions.
     Results Showing: Ultraviolet absorption spectrum did not change with theextension of radiation time, the irradiation time when the radiation intensity of8×100μJ/cm~2to5T (20s), the UV absorption spectrum changed significantly, i.e. thecharacteristic peaks disappear; radiation time to continue to extend the radiationintensity, the UV absorption spectrum does not change. Irradiation time (20s)5TSynthesis of Az-G photoactive gelatin crosslinking reaction can occur in the radiationintensity of8×100μJ/cm~2. In accordance with this crosslinking reaction conditionsfurther analysis of the UV absorption spectrum before and after the change, and theultraviolet absorption spectrum of the ultraviolet radiation before Az-G photoactivegelatin obvious peak at268.4nm; when the radiation intensity reaches8x100μJ/cm~2, compared with ultraviolet radiation before the radiation time of20s, thephotoactive radiation gelatin UV absorption spectrum at268.4nm at the crestdisappear. Therefore, it is considered that the photoactive gelatin the optimumcrosslinking reaction conditions: radiation intensity is8×100μJ/cm~2radiation timeof20s.
     3. The Az-G photoactive gelatin in the fixed effect of the type of materialand the surface of the stent
     First fixed plates table light modification: After drying, ultraviolet radiation,washing and dyeing, Az-G photoactive gelatin is fixed to the inner surface of theculture plate and dyed blue Reference Synthesis of Az-G light active gelatin with lightactivity, and fixed through fixed optical technology in the surface of the culture plate;Subsequently,200mL of PBS buffer added to the light fixed hole of the96-wellplates, were incubated with3,7days in an incubator at37°C, using Coomassie bluedye and found that the culture plate surface is still clearly visible circular blue spotsfixed AZ-G the photoactive gelatin and synthetic AZ-G photoactive Descriptiongelatin was dissolved over time, light fixed with excellent results, will not be affectedby the effects of temperature and solution off, and stable up to7days. In contrast,pure gelatin to be washed off after the ultraviolet radiation, the hole is not formed blueround spots, gelatin alone can not be fixed in the surface of the culture plate.
     Using the method of fixing the light modification experiments carried out in thetype of polymer film and the surface of the stent. After drying, ultraviolet radiation,washed, stained, Az-G photoactive gelatin either in PLGA and PLA, and PVA filmsurface, or in the the PLGA electrospinning film and PVA hydrogel stent surface, areformed a visible blue circular spot color, and pure gelatin did not leave visible tracesof blue on the polymer surface. Technology by light fixed to achieve photoactivegelatin PLGA, PLA, and PVA films and PLGA electrostatic spinning membrane andPVA hydrogel surface of the stent fixation also demonstrated the synthesis ofoptically active gelatin having optical activity.
     二、Synthesis of new materials for skin tissue engineering and Cytological studieson its modificated surface
     1. Preparation of electrospinning PLGA fiber membrane
     In this study, prepared by electrospinning PLGA electrospun membrane, theelectrospinning fibers of uniform thickness, diameter in the nanometer range, betweenthe fibers irregular arrangement.
     2. Preparation of PVA hydrogel by Frozen–thawed method
     PVA hydrogel prepared by repeated freezing, the present study was visually observed as a white gelatinous scanning electron microscope PVA hydrogel internalpore structure having a dense, pore sizes, shapes, irregular, the space lattice-shapedstructure is a tissue engineering scaffolds.
     3. Fabrication and characterization of double artificial scaffolds of PLGA/PVA
     Electrospinning PLGA film and PVA hydrogel as Artificial dressing defects, thisstudy combines the advantages of both, and the use of physical crosslinking way bothcomposite construct a similar double skin structure artificial bracket.
     Mechanical properties test results show that, compared with the pure PVAhydrogel scaffolds, composite PLGA electrospun membrane of PLGA/PVA doublestent to the superior mechanical properties of PLGA/PVA double stent to stretchstrength, elastic modulus, up to the point of breaking force energy, tensile strengthstrain, tensile breaking strain gauge tensile maximum load were higher than thesingle-layer PVA hydrogel, and the difference was statistically significant (p <0.05);PLGA/PVA double artificial support compared with the single-layer PVA hydrogeltensile stress at break and tensile breaking load were not statistically significant (P>0.05), but have a similar trend, the PLGA/PVA double stent to The tensile breakingstress and tensile breaking load were higher than the single layer of PVA hydrogel.
     Determination of PLGA/PVA double artificial bracket and the moisture contentof the PVA hydrogel were81.95±1.880and83.93±3.262, statistical analysisshowed no difference (p>0.05) between. Double dressing and different Time PLGA/PVA PVA hydrogel dehydration rate is not the same, the two dressings in the initialstage of dehydration dehydration rate gradually increases slower dewatering rate is therate of change with time, until constant weight no longer changes; constant weight of135min dehydrated PVA hydrogel of not change thereafter dewatering rate, PLGA/PVA double stent to constant weight is reached only at315min after dehydration; thesame dehydration time after, two dressing dehydration rate is significantly differentPVA hydrogel initially dewatering rate was significantly greater than the PLGA/PVA Duplex dressing (p <0.05), with time, when the the dehydration time reaches 315min, both dehydration rate No statistically significant difference (p>0.05).Description of the the double stent to physically prepared not only has the advantageof rich moisture PVA hydrogel containing PLGA electrospinning layer of the surfacecan stop the rapid loss of moisture, delaying the drying time of the PVA hydrogel,play water retention effect.
     4. Preparation of patterned PVA hydrogel
     The present study was prepared by the mold SYSTEM "patterned" PVAhydrogel, the method to change the structure of the hydrogel surface, rough stripedsurface is conducive to cell attachment and growth,"porous" PVA hydrogel formedon the surface narrow pore structure is conducive to early adhesion of cells.
     5. The Surface Modification of Biological Basic Research on skin tissueengineering scaffold by Az-G photoactive gelatin
     The a study prepared AZ-G photoactive gelatin PLGA film, PVA hydrogelsurface modification, the modified surface can promote the adhesion of L929cells toaccelerate the rate of the adherent cells, and with the time and the photoactive gelatinconcentration increases, cell spreading, growth and proliferation capacity graduallyincreases, without modification of the number of parts of the cell, and the adhesion,growth and proliferation capacity is much lower than the modified portion; thephotoactive gelatin PLGA the electrospinning surface modified did not affectfibroblast adhesion rate, stretch, growth and proliferation of cells had no significantrole in promoting; photoactive gelatin Cytological studies on different tissueengineering scaffold material surface modification that The use concentration of thegelatin of the present study was prepared AZ-G photoactive at0.1mg/mL, the materialsurface can be modified to achieve the purpose of promoting cell adhesion, growthand proliferation.
     Conclusions: In this study, the photoactive azido group and the gelatin moleculescarboxy occurrence of a condensation reaction, as having photoactive newphotoactive compound-AZ-G is formed gelatin; through the anchoring effect of thesurface of the material and cytologic evaluation proved Az-G light active gelatin stable fixed to the surface of the biological material, will not fall off, and the cellcompatibility, is conducive to cell adhesion, growth and proliferation; This is the firstthe electrospinning film and PVA hydrogel by Acta frozen crosslinked methodcombines the performance of their models and water retention is much better than asimple the PLGA the electrospinning film or PVA hydrogel; mold prepared "porous"PVA hydrogel is conducive to cell adhesion and Growth.
引文
[1] Bisceglie V. Uber die antineoplastische immunitat; heterologe Einpflanzungvon Tumoren in Huhner-embryonen [J]. Ztschr. Krebsforsch,1993,40:122–140.
    [2] Chick, W. L., Like, A. A., Lauris, V. Beta cell culture on synthetic capillaries:An artificial endocrine pancreas [J]. Science,1975,187:847–848.
    [3] Bell, E., Ehrlich, P., Buttle, D. J., Nakatsuji, T. Living tissue formed in vitroand accepted as skin-equivalent of full thickness [J]. Science,1981,221:1052–1054.
    [4] Burke, J. F., Yannas, I. V., Quimby, W. C., et a1. Successful use of aphysiologically acceptable artificial skin in the treatment of extensive burninjury [J]. Ann Surg,1981,194:413–448.
    [5] Nerem RM. Tissue engineering in the USA [J]. Med Biol Eng Comput,1992,30(4):CE8-CE12.
    [6] Langer R, Vacanti JP. Tissue engineering [J]. Science,1993,260:920-926.
    [7] MS Chapekar. Tissue engineering: Challenges and opportunities [J]. Journalof Biomedical Materials Research,2000,53(6):617-620.
    [8] Goldstein S. Presentation. Cambridge Health Tech Institute’s2nd annual tissueengineering/regenerative healing/stem cell biology meeting, Pittsburg,October3-5,1999[C].USA,c1999.
    [9] Niklason LE, Gao J, Abbott WM, et a1. Functional arteries grown in vitro [J].Sci,1999,284:489–493.
    [10] Huynh T, Abraham G, Murray J, et a1. Remodeling of an acellular collagengraft into a physiologically responsive neovessel [J]. Nat Biotechnol,1999,17:1083–1086.
    [11] Lanza R, Chick WL. Bioartificial organs: Science, medicine, and technology[J]. Ann NY Acad Sci,1997,831:323–331.
    [12] Chen SC, Mullon C, Kahaku E, et a1. Bioartificial organs: Science, medicine,and technology [J]. Ann NY Acad Sc,1997;831:350–360.
    [13] Oberpenning F, Meng J, Yoo JJ, et a1. De novo reconstitution of a functionalmammalian urinary bladder by tissue engineering [J]. Nat Biotechnol,1999,17:149–155.
    [14] Humes HD, Buffington DA, Mackay SM, et a1. Replacement of renalfunction in uremic animals with a tissue-engineered kidney [J]. NatBiotechnol,1999,17:451–455.
    [15] David F. Muir, Gainesville, FL. Materials and Methods for Nerve Repair: US,12/966,540[P].2010-12-13.
    [16] Jin-Seok Yi, M.D., Hyung-Jin Lee, M.D., Hong-Jae Lee, M.D., et al. RatPeripheral Nerve Regeneration Using Nerve Guidance Channel by PorcineSmall Intestinal Submucosa [J]. J Korean Neurosurg Soc,2013,53(2):65-71.
    [17] Blight AR. Cambridge HealthTech Institute’s2nd annual tissueengineering/regenerative healing/stem cell biology meeting [C]. Pittsburgh,Oct3–5,1999.
    [18] Xu XM, Zhang SX, Li H, et al. Fetal spinal cord tissue in mini-guidancechannels promotes longitudinal axonal growth after grafting into hemisectedadult rat spinal cords [J]. Neural Plasticity,1999,6(4):103–121.
    [19] Hadlock T, Singh S, Vacanti JP, McLaughlin BJ. Ocular cell monolayerscultured on biodegradable substrates [J]. Tissue Eng,1999,5:187–196.
    [20] Germain L, Auger FA, Grandbois E, et al. Reconstructed human corneaproduced in vitro by tissue engineering [J]. Pathobiol,1999,67:140–147.
    [21] Nishi K, Mano C, Ichihara M, Honda T, et al. The inhibition of lens epithelialcell migration by a discontinuous capsular bend created by aband-shapedcircular loop or a capsule-bending ring [J]. Ophthalmic Surg Lasers,1998,29:119–125.
    [22] Awad HA, Butler DL, Boivin GP, et al. Autologous mesenchymal stemcell-mediated repair of tendon [J]. Tissue Eng,1999,5:267–277.
    [23] Cao Y, Vacanti JP, Ma X, et al. Generation of neotendon using syntheticpolymers seeded with tenocytes [J]. Transplant Proc,1995,26:3390–3392.
    [24] Stading M, Langer R. Mechanical shear properties of cell-polymer cartilageconstructs [J]. Tissue Eng Tissue Eng,1999,5:241–250.
    [25] Schreiber RE, Ilten–Kirby BM, Dunkelman NS, et al. Repair of osteochondraldefects with allogeneic tissue engineered cartilage implants [J]. Clin OrthopRelat Res,1999,367S:382–395.
    [26] Huang D, Chang TR, Aggarwal A, et al. Mechanianims and dynamics ofmechanical strengthening in ligament equivilent fibroblast populated collagenmatrices [J]. Ann Biomed Eng,1993,21:289–305.
    [27] Isogai N, Landis W, Kim TH, et al. Formation of phalanges and small jointsby tissue-engineering. J Bone Joint Surg81A:306–316[J]. J Bone Joint Surg,1999,81A:306–316.
    [28] Bohi KS, Shon J, Rutherford B, et al. Role of synthetic extracellularmatrix in development of engineered dental pulp [J]. J Biomater Sci Polym Ed,1998,9:749–764.
    [29] Black AF, Berthod F, L’Heureux N, et al. In vitro reconstruction of a humancapillary-like network in a tissue-engineered skin equivalent [J]. FASEB J,1998,12:1331–1340.
    [30] Tabata, Y. The importance of drug delivery systems in tissue engineering [J].Pharm. Sci. Technol.,2000,3:80–89.
    [31] P Ferretti, J Géraudie. Cellular and Molecular Basis of Regeneration: FromInvertebrates to Humans [M]. Chichester: John Wiley&Sons Ltd,1997.
    [32] Shin-oka T, Shum-Tim D, MaP, et al.1997. Tissue-engineered heart valveleaflets—Does cell origin affect outcome? Circulation96(Suppl. II): II-102–7.
    [33] R. A. Pollak, H. Edington, J. L. Jensen, et al. A human dermal replacementfor the treatment of diabetic foot ulcers [J]. Wounds,1997,9:175-183.
    [34] Edmond A. Ryan, Jonathan R.T. Lakey, Ray V. Rajotte, et al. ClinicalOutcomes and Insulin Secretion After Islet Transplantation With theEdmonton Protocol [J]. Diabetes,2001,50(4):710-719.
    [35] Strom SC, Roy-Chowdhury J, Fox IJ. Hepatocyte transplantation for thetreatment of human disease [J]. Sem Liver Dis,1999:19:39–48.
    [36] J. Mansbridge, K. Liu, R. Patch, et al. Three-dimensional fibroblast cultureimplant for the treatment of diabetic foot ulcers: metabolic activity andtherapeutic range [J]. Tissue Eng,1998,4:430.
    [37] A. Kern, K. Liu, J. Mansbridge, J. Modification of fibroblast γ-interferonresponses by extracellular matrix [J]. Invest Dermatol,2001,117:112-118.
    [38]杨照,周新民,樊代明,等.生物型人工肝的新种子细胞来源-诱导性多潜能干细胞[J].胃肠病学和肝病学杂志,2010,19(3):193-195.
    [39] A. J. Strain, J. M. Neuberger, A bioartificial liver--state of the art [J]. Science,2002,295:1005-1009.
    [40] Thomson JA, Itskovitz-Eldor J, Shapiro SS, et al. Embryonic stem cell linesderived from human blastocysts [J]. Science,1998,282:1145-7.
    [41] Potten, C.S. Stem Cells [M]. London:Academic Press Ltd, Harcourt Brace&Company,1997.
    [42] Thomson JA, Itskovitz-Eldor J, Shapiro SS, et al. Embryonic stem cell linesderived from human blastocysts [J]. Science,1998,252:1145–47.
    [43] Cerqueora MT, Marques AP, Reis RL. Using stem cells in skin regeneration:possibilities and reality [J]. Stem Cells Dev,2012,21(8):1201-1214.
    [44] Vincent Falanga. Stem Cells in Tissue Repair and Regeneration [J]. J InvestDermatolo,2012,132:1538-1541.
    [45] Evans MJ, Kaufman MH. Establishment in culture of pluripotential cells frommouse embryos [J]. Nature,1981,292(5819):154-156.
    [46] Martin GR. Isolation of a pluripotent cell line from early mouse embryoscultured in medium conditioned by teratocarcinoma stem cells [J].Proceedings of the National Academy of Sciences,1981,78(12):7634-7638.
    [47] Nourse MB, Halpin DE, Scatena M, et al. VEGF Induces differentiation offunctional endothelium from human embryonic stem cells implications fortissue engineering [J]. Arteriosclerosis Thrombosis and Vascular Biology,2010,30(1):80-89.
    [48] Li Zongjin, Suzuki Y, Huang Mei, et al. Comparison of reporter gene and ironparticle labeling for tracking fate of human embryonic stem cells anddifferentiated endothelial cells in living subjects [J]. Stem Cells,2008,26(4):864-873.
    [49] Xie Changqing, Zhang Jifeng, Villacorta L, et al. A highly efficient method todifferentiate smooth muscle cells from human embryonic stem cells [J].Arteriosclerosis Thrombosis and Vascular Biology,2007,27(12):1311-1312.
    [50] Bai Hao, Gao Yongxing, Arzigian M, et al. BMP4regulates vascularprogenitor development in human embryonic stem cells through asmad-dependent pathway [J]. Journal of Cellular Biochemistry,2009,109(2):363-374.
    [51] James D, Nam H, Seandel M, et al. Expansion and maintenance of humanembryonic stem cell-derived endothelial cells by TGFβ inhibition is Id1dependent [J]. Nature Biotechnology,2010,28(2):161-166.
    [52] Ferreira LS, Gerecht S, Shieh HF, et al. Vascular Progenitor Cells IsolatedFrom Human Embryonic Stem Cells Give Rise to Endothelial and SmoothMuscle-Like Cells and Form Vascular Networks In Vivo [J]. CirculationResearch,2007,101(3):286-294.
    [53] Pepe AE, Xiao Qingzhong, Zampetaki A, et al. Crucial role of nrf3in smoothmuscle cell differentiation from stem cells [J]. Circulation Research,2010,106(5):870-879.
    [54] Blancas AA, Shih AJ, Lauer NE, et al. Endothelial cells from embryonic stemcells in a chemically defined medium [J]. Stem Cells and Development,2011,20(12):2153-2161.
    [55] Descamps B, Emanueli C. Vascular differentiation from embryonic stem cells:Novel technologies and therapeutic promises [J]. Vascular Pharmacology,2012,56(3):267-279.
    [56] Kane NM, Meloni M, Spencer HL, et al. Derivation of endothelial cells fromhuman embryonic stem cells by directed differentiation analysis of microRNAand angiogenesis In vitro and in vivo [J]. Arteriosclerosis Thrombosis andVascular Biology,2010,30(7):1389-1397.
    [57] Matsuura K, Masuda S, Haraguchi Y, et al. Creation of mouse embryonicstem cell-derived cardiac cell sheets [J]. Biomaterials,2011,32(30):7355-7362.
    [58] Willems E, Cabral-Teixeira J, Schade D, et al. Small moleculemediated TGFβtype II receptor degradation promotes cardiomyogenesis in embryonic stemcells [J]. Cell Stem Cell,2012,11(2):242-252.
    [59] Ahmad S, Stewart R, Yung Sun, et al. Differentiation of human embryonicstem cells into corneal epithelial-like cells by in vitro replication of thecorneal epithelial stem cell niche [J]. Stem Cells,2007,25(5):1145-1155.
    [60] Akanuma H, Qin Xianyang, Nagano R, et al. Identification of stage-specificgene expression signatures in response to retinoic acid during the neuraldifferentiation of mouse embryonic stem cells [J]. Frontiers in Genetics,2012,141(3):1-12.
    [61] Toh WS, Lee EH, Cao Tong. Potential of human embryonic stem cells incartilage tissue engineering and regenerative medicine [J]. Stem Cell Reviewsand Reports,2011,7(3):544-559.
    [62] Shi DL. Introduction to biomaterials [M]. Beijing: Tsing hua University Press,2006.
    [63] Hutmacher D W. Scaffold design and fabrication technologies for engineeringtissues state of the art and future perspectives [J]. J Biomater Sci Polym Ed,2001,12(1):107.
    [64] Hoppe A, Guldal N S, Boccaccini A R. A review of the biological response toionic dissolution products from bioactive glasses and glass-ceramics [J].Biomaterials,2011,32(11):2757.
    [65] YU Ting, LIU Ya, WANG Yu, et al. Preparation and Bioactivity of theComposite of PLGA and Hydroxyapatite Nanocrystals Surface-grafted withL-lactic Acid Oligomer [J]. Chem. J. Chinese Universities,2009,30(7):1439-1444.
    [66] DONG Shu-Jun, YU Ting, WEI Jun-Chao, et al. Preparation SurfaceProperties and Biological Activity of the Composite ofPoly(lactide-co-glycolide) and Bioglass Nanoparticles Surface Grafted withPoly(L-lactide)[J]. Chem. J. Chinese Universities,2009,30(7):1439-1444.
    [67] WANG Lin, LI Chun-Yan, HE Pan, et al. Preparation and Bioactivities ofPLGA/Nano-hydroxyapatite Scaffold Containing Chitosan Microspheres forControlled Delivery of Mutifuncational Peptide-adrenomedullin [J]. Chem. J.Chinese Universities,2011,32(7):1622-1628.
    [68] CUI Wei-Wei, LIU Ya, WANG Zong-Liang, et al. Preparation and BiologicalEvaluation of Electrospun MSM/PLGA Dressing Containing Nano-silver [J].Chem. J. Chinese Universities,2013,34(3):679-685.
    [69] Narayan R. Biomedical materials [M]. New York:Springer,2009.
    [70] Rita Levi-Montalcini, Stanley Cohen. Effects of the extract of the mousesubmaxillary salivary glands on the sympathetic system of mammals [J]. AnnNY Acad Sci,1960,85(3):324-341.
    [71] Yasuhiko Tabata. The importance of drug delivery systems in tissueengineering [J]. Pharm. Sci. Technol,2000,3:80-89.
    [72] Fontaine, M. J., Hansen, L. K., Thompson, S., et al. Transplantation ofgenetically altered hepatocytes using cell–polymer constructs leads tosustained human growth hormone secretion in vivo [J]. Transplant. Proc.,1993,25:1002–1004.
    [73] Hanpanich, B.S. Tissue engineering of skin and soft tissue augmentation,medical view [J]. J Med Assoc Thai,2010,93:7.S332-6.
    [74] Groeber, F., Holeiter M., Hampel M., et al. Skin tissue engineering in vivoand in vitro applications [J]. Adv Drug Deliv Rev,2011,63:352-366.
    [75] Danisovic, L., Varga I., Zamborsky R.,et al. The tissue engineering ofarticular cartilage: cells, scaffolds and stimulating factors [J]. Exp Biol Med,2012,237(1):10-17.
    [76]吴先林.皮肤性病学[M].北京:人民卫生出版社,2001.
    [77]张学军,刘维达,何春涤.现代皮肤病学基础[M].北京:人民卫生出版社,2010.
    [78] Alexandra C. Kendall, Anna Nicolaou. Bioactive lipid mediators in skininflammation and immunity [J]. Progress in Lipid Research,2013,52(1):141–164.
    [79] Westerhof W. The discovery of the human melanocyte [J]. Pigment Cell Res,2006,19(3):183-193.
    [80] Romani N., Brunner P.M., Stingl G. Changing views of the role ofLangerhans cells [J]. J Invest Dermatol,2012,132:872-81.
    [81] Polakovicova S., Seidenberg H., Mikusova R., et al. Merkel cells--review ondevelopmental, functional and clinical aspect [J]. Bratisl Lek Listy,2011,112(2):80-87.
    [82] Stadelmann W.K., Digenis A.G., Tobin G.R. Impediments to wound healing[J]. Am J Surg,1998,176(2):39S-47S.
    [83] Clark, R.A. Potential roles of fibronectin in cutaneous wound repair [J]. ArchDermatol,1988,124(2):201-206.
    [84] Shaw T.J., Kishi K., Mori R. Wound-associated skin fibrosis: mechanisms andtreatments based on modulating the inflammatory response [J]. Endocr MetabImmune Disord Drug Targets,2010,10(4):320-330.
    [85] Mahdavian Delavary B., van der Veer W.M., van Egmond M., et al.Macrophages in skin injury and repair [J]. Immunobiology,2011,216(7):753-762.
    [86] Bittencourt FV, Marghoob AA, Kopf AW, et al. Large congenital melanocyticnevi and the risk of development of malignant melanoma and neurocutaneousmelanocytosis [J]. Pediatrics,2000,106:736–741.
    [87] Zaal LH, Mooi WJ, Sillevis Smitt JH, et al. Classification of congenitalmelanocytic naevi and malignant transformation: a review of the literature [J].Br J Plast Surg,2004,57:707–719.
    [88] Bauer BS, Corcoran J. Treatment of large and giant nevi [J]. Clin Plast Surg,2005,32:11–18.
    [89] Earle SA, Marshall DM. Management of giant congenital nevi with artificialskin substitutes in children [J]. J Craniofac Surg,2005,16:904–907.
    [90] Tannous ZS, Mihm Jr MC, Sober AJ, Duncan LM. Congenital melanocyticnevi: clinical and histopathologic features, risk of melanoma, and clinicalmanagement [J]. J Am Acad Dermatol,2005,52:197–203.
    [91] Arneja JS, Gosain AK. Giant congenital melanocytic nevi of the eyelids andperiorbital region [J]. Plast Reconstr Surg,2007,120:26e–40e.
    [92] Kryger ZB, Bauer BS. Surgical management of large and giant congenitalpigmented nevi of the lower extremity [J]. Plast Reconstr Surg,2008,121:1674–84.
    [93] Schiestl C, Stiefel D, Meuli M. Giant naevus, giant excision, eleg(i)antclosure? Reconstructive surgery with Integra Artificial Skin1to treat giantcongenital melanocytic naevi in children [J]. J Plast Reconstr Aesthet Surg,2010,63(4):610-615.
    [94] Sophie Bo¨ttcher-Haberzeth, Thomas Biedermann, Ernst Reichmann. Tissueengineering of skin [J]. Burns,2010,36:450–460.
    [95] Aarabi S, Longaker MT, Gurtner GC. Hypertrophic scar formation followingburns and trauma: new approaches to treatment [J]. PLoS Med,2007,4:e234.
    [96] Berman B, Viera MH, Amini S, et al. Prevention and management ofhypertrophic scars and keloids after burns in children [J]. J Craniofac Surg,2008,19:989–1006.
    [97] Karasek M. In vitro culture of human skin epithelial cell [J]. J Invest Dermatol,1966,47:533–40.
    [98] Billingham RE, Medawar P.Technique of free skin grafting in mammals [J]. JExp Biol,1950,28:385–402.
    [99] Rheinwald JG, Green H. Epidermal growth factor and the ultiplication ofcultured human pidermal eratinocytes [J]. Nature,1977,265:421–4.
    [100] Rheinwald JG, Green H. Serial cultivation of strains of uman epidermalkeratinocytes: the formation of eratinizing colonies from single cells [J]. Cell,1975,6:331–44.
    [101] O’Connor N, Mulliken JB, Banks-Schlegel S, et al. Grafting of burns withcultured epithelium repared from autologous epidermal cells [J]. Lancet,1981,1:75–8.
    [102] Gallico GG, O’Connor NE, Compton CC, et al. Permanent coverage of largeburn wounds with autologous cultured human epithelium [J]. N Engl J Med,1984,311:448–51.
    [103] Cuono C, Langdon R, McGuire J. Use of cultured epidermal autografts anddermal allografts as skin replacement after burn injury [J]. Lancet,1986,17:1123–4.
    [104] Munster AM. Cultured skin for massive burns-A prospective, controlled trial[J]. Ann Surg,1996,224:372–5.
    [105] Carsin H, Ainaud P, Le Bever H, et al. Cultured epithelial autografts inextensive burn coverage of severely traumatized patients: a five yearsingle-center experience with30patients [J]. Burns,2000,26:379–87.
    [106] Ronfard V, Rives JM, Neveux Y, et al. Long-term regeneration of humanepidermis on third degree burns transplanted with autologous culturedepithelium grown on a fibrin matrix [J]. Transplantation,2000,70:1588–98.
    [107] Hernon CA, Dawson RA, Freedlander E, et al. Clinical experience usingcultured epithelial autografts lead to an alternative methodology fortransferring skin cells from the laboratory to the patient [J]. Regen Med,2006,1:809–21.
    [108] Wood FM, Kolybaba ML, Allen P. The use of cultured epithelial autograft inthe treatment of major burn wounds: eleven years of clinical experience [J].Burns,2006,32:538–44.
    [109] Atiyeh BS, Costagliola M. Cultured epithelial autograft (CEA) in burntreatment: three decades later [J]. Burns,2007,33:405–13.
    [110] Herndon DN, Parks DH. Comparison of serial debridement and autograftingand early massive excision with cadaver skin overlay in the treatment of largeburns in children [J]. J Trauma,1986,26:149–52.
    [111] Barrandon Y, Green H. Three clonal types of keratinocyte with differentcapacities for multiplication [J]. Proc Natl Acad Sci,1987,84:2302–2306.
    [112] Mavilio F, Pellegrini G, Ferrari S, et al. Correction of junctionalepidermolysis bullosa by transplantation of genetically modified epidermalstem cells [J]. Nat Med,2006,12:1397–402.
    [113] Rue LW, Cioffi WG, McManus WF, et al. Wound closure and outcome inextensively burned patients treated with cultured autologous keratinocytes [J].J Trauma,1993,34:662–667.
    [114] Bell E, Ehrlich HP, Buttle DJ, et al. Living tissue formed in vitro and acceptedas skin-equivalent tissue of full thickness [J]. Science,1981,211:1052–1054.
    [115] Boyce ST, Goretsky MJ, Greenhalgh DG, et al. Comparative assessment ofcultured skin substitutes and native skin autograft for treatment offullthickness burns [J]. Ann Surg,1995,222:743–752.
    [116] Delvoye P, Pierard D, Noel A, et al. Fibroblasts induce the assembly of themacromolecules of the basement membrane [J]. J Invest Dermatol,1988,90:276–82..
    [117] Ko¨nig A, Bruckner-Tuderman L. Epithelial-mesenchymal interactionsenhance expression of collagen VII in vitro [J]. J Invest Dermatol,1991,96:803–808.
    [118] Burke JF, Yannas IV, Quinby Jr WC, et al. Successful use of aphysiologically acceptable artificial skin in the treatment of extensive burninjury [J]. Ann Surg,1981,194:413–428.
    [119] Sheridan RL, Tompkins RG. Skin substitutes in burns [J]. Burns,1999,25:97–103.
    [120] Pandya AN, Woodward B, Parkhouse N. The use of cultured autologouskeratinocytes with Integra1in the resurfacing of acute burns [J]. PlastReconstr Surg,1998,102:825–828.
    [121] EA Woodroof. The search for an ideal temporary skin substitute: AWBAT [J].Eplasty,2009,12(2):9:e10.
    [122] Pruit BA Jr, Levine NS. Charaterstics and use of biologic dressing and skinsubstitutes [J]. Arch Surg,1984,119:312-322.
    [123] Wainwright DJ. Use of an acellular allograft dermal matrix(Alloderm)in themanagement of full-thickness burns [J]. Burns,1995,21:243-248.
    [124] Larouche D, Paquet C, Fradette J, et al. Regeneration of skin and cornea bytissue engineering [J]. Methods Mol Biol,2009,482:233-256.
    [125] Kalyanaraman B, Supp DM, Boyce ST. Medium flow rate regulates viabilityand barrier function of engineered skin substitutes in perfusion culture [J].Tissue Eng Part A,2008,14:583-593.
    [126] Hernon CA, Harrison CA, Thornton DJ, et al. Enhancement of keratinocyteperformance in the production of tissueengineered skin using a low-calciummedium [J]. Wound Repair Regen,2007,15:718-726.
    [127] Hu S, Kirsner RS, Falanga V, et al. Evaluation of Apligraf persistence andbasement membrane restoration in donor site wounds: a pilot study [J].Wound Repair Regen,2006,14:427-433.
    [128] Li-Ling Tang, Huan Liu, Yuan-Liang Wang, et al. Evaluation of thebiocompatibility of acellular porcine dermis [J]. Colloids and Surfaces B:Biointerfaces,2007,57(2):215–218.
    [129] Liangpeng Ge, Shuquan Zheng, Hong Wei. Comparison of histologicalstructure and biocompatibility between human acellular dermal matrix (ADM)and porcine ADM [J]. Burns,2009,35(1):46–50.
    [130] Chavarriaga Luis Felipe, Lin Edward, Losken Albert, et al. Management ofComplex Abdominal Wall Defects Using Acellular Porcine Dermal Collagen[J]. The American Surgeon,2010,76(1):96-100.
    [131] I.Himsl, V. Drinovac, M. Lenhard, et al. The use of porcine acellular dermalmatrix in silicone implant-based breast reconstruction [J]. Archives ofGynecology and Obstetrics,2012,286(1):187-192.
    [132] Jiong Chen, Jiake Chai, Chunmao Han, et al. Clinical Application andLong-Term Follow-Up Study of Porcine Acellular Dermal Matrix CombinedWith Autoskin Grafting [J]. Journal of Burn Care&Research,2010,31(2):280-285.
    [133]沈光裕,潘银根,陈建设,等.猪脱细胞真皮基质在人皮肤创伤修复中的临床应用[J].华南国防医学杂志,2010,2:123-125.
    [134] Xiangsheng Feng, Jiaju Tan, Yingen Pan, et al. Control of hypertrophic scarfrom inception by using xenogenic (porcine) acellular dermal matrix (ADM)to cover deep second degree burn [J]. Burns,2006,32(3):293–298.
    [135]黄伟琪,赵耀华,邓海涛,等.基因转染猪皮在深度烧伤创面治疗中的应用[J].中华损伤与修复杂志,2012,4:41-42.
    [136]邹勃生,谭军,李效,等.青蛙皮及乳猪皮治疗深度烧伤创面的比较研究[J].中国医师杂志,2004,6(1):74-76.
    [137] Gobssler-Schobfberger R, Hesser G, Muik M, et al. An orphan dermaseptinfrom frog skin reversibly assembles toamyloid-like aggregates in apH-dependent fashion [J]. FEBS J,2009,276(20):5849-5859.
    [138] Kest ing M R, Wo lff K D, Hohlw eg-Majert B, et al. The r ole of allo genicamnio tic membra ne in burn tr eatment [J]. J Bur n Care Res,2008,29(6):907-916.
    [139]庞建华,杨嵩峰,陈伟华,等.复层辐射人羊膜应用于烧伤创面的研究[J].中华整形烧伤外科杂志,2004,4(2):22-26.
    [140] Sheila MacNeil. Biomaterials for tissue engineering of skin [J].Materialstoday,2008,11(5):26–35.
    [141] Catherine A Hernon, Rebecca A Dawson, Eric Freedlander, et al. Clinicalexperience using cultured epithelial autografts leads to an alternativemethodology for transferring skin cells from the laboratory to the patient [J].Regenerative Med,2006,6(11):809-821.
    [142] Phillips TJ. New skin for old: developments in biological skin substitutes [J].Arch Dermatol,1998,134(3):344-349.
    [143] F. Berthod, O. Damour. In vitro reconstructed skin models for woundcoverage in deep burns [J]. Br J Dermatol,1997,136(6):809-816.
    [144] Wood FM, Kolybaba ML, Allen P. The use of cultured epithelial autograft inthe treatment of major burn injuries: a critical review of the literature [J].Burns,2006,32(4):395-401.
    [145] Zhang JY, Wang CM. Research and progress in tissue-engieered skin [J].Journal of Clinical Rehabilitative Tissue Engineering Research,2007,11(10):1988-1991.
    [146] Gilcrest BA,Calhoun JK,Maciag T. Attachment and growth of humankeratinocytes in a serum-free environment [J]. J Cell Physiol.,1982,112:197-206.
    [147] Caissie R, Gingras M, Champigny MF, et al. In vivo enhancement of sensoryperception recovery in tissue-engineered skin enriched with laminin [J].Biomaterials,2006,27(15):2988-2993.
    [148] Hetton JM, Madden MR, Finkelstein JL, et al. Grafting of burn patients withallografts of cultured epidermal cells [J]. Lancet,1983,11:428-430.
    [149] Gielen V, Faure M, Maudiut G, et al. Progressive replacement of humancultured epithelial allografts by recipient cells as evidenced by HLA class Iantigens expression [J]. Dermatologica,1987,175:166-170.
    [150] Cuono C, Langdon R, McGuire J. Use of cultured epidermal autografts anddermal allografts as skin replacement after burn injury [J]. Lancet,1986,1:1123–1124.
    [151] Burke JF, Yannas IV, Quinby WC Jr, et al. Successful use of aphysiologically acceptable artificial skin in the treatment of extensive burninjury [J]. Ann Surg,1981,194:413–428.
    [152] Heimbach D, Luterman A, Burke J, et al. Artificial dermis for major burns: amulticenter randomized clinical trial [J]. Ann Surg,1988,208:313–20.
    [153] Purdue GF. Dermagraft-TC pivotal safety and efficacy study [J]. J Burn CareRehabil,1997,18:S13–14.
    [154] Gentzkow GD, Iwasaki SD, Hershon KS, et al. Use of dermagraft: a culturedhuman dermis to treat diabetic foot ulcers [J]. Diabetes Care,1996,19:350–354.
    [155] Marston WA, Hanft J, Norwood P, et al. The efficacy and safety ofDermagraft in improving the healing of chronic diabetic foot ulcers: results ofa prospective randomized trial [J]. Diabetes Care,2003,26:1701–1705.
    [156] Ehrenreich M, Ruszczak Z. Update on tissue-engineered biological dressings[J]. Tissue Eng,2006,12:1–18.
    [157] Bouchle A. Tissue engineering firms go under [J]. Nat Biotechnol,2002,20:1178–9.
    [158] Mostow EN, Haraway GD, Dalsing M, et al. Effectiveness of an extracellularmatrix graft (OASIS Wound Matrix) in the treatment of chronic leg ulcers: arandomized clinical trial [J]. J Vasc Surg,2005,41:837–843.
    [159] Cole P, Hom TW, Thaller S. The use of decellularized dermal grafting(AlloDerm) in persistent oro-nasal fistulas after teriary cleft palate repair [J]. JCraniofac Surg,2006,17(4):636-641.
    [160] Callcut RA, Schurr MJ, Sloan M, et al. Clinical experience with alloderm: aone-staged composite dermal/epidermal replacement utilizing processedcadaver dermis and thin autografts [J]. Burn,2006,32(5):583-588.
    [161] DN HERNDON, DH PARKS. Comparison of Serial Debridement andAutografting and Early Massive Excision with Cadaver Skin Overlay in theTreatment of Large Burns in Children [J]. Trauma,1986,26(2):149.
    [162] D N Herndon, R E Barrow, R L Rutan, et al. A comparison of conservativeversus early excision(Therapies in severely burned patients)[J]. Ann Surg,1989,209(5):547-553.
    [163] J F Burke, I V Yannas, W C Quinby, et al. Successful use of a physiologicallyacceptable artificial skin in the treatment of extensive burn injury [J]. Ann.Surg.,1981,194:413-428.
    [164] Bell E, Ivarsson B, Merrill C. Production of a tissue-like structure bycontraction of collagen lattices by human fibroblasts of different proliferativepotential in vitro [J]. Proc Natl Acad Sci,1979,76:1274–1278.
    [165] Bell E, Ehrlich HP, Buttle DJ, et al. Living tissue formed in vitro and acceptedas skin-equivalent tissue of full thickness [J]. Science,1981,211:1052–1054.
    [166] Eaglstein WH, Iriondo M, Laszlo K. A composite skin substitute (graftskin)for surgical wounds: a clinical experience [J]. Dermatol Surg,1995,21:839–843.
    [167] Sabolinski ML, Alverez O, Auletta M, et al. Cultured skin as a ‘smartmaterial’ for healing wounds: experience in venous ulcers [J]. Biomaterials,1996,17:311–320.
    [168] Falanga V, Margolis D, Alvarez O, et al. Rapid healing of venous ulcers andlack of clinical rejection with an allogeneic cultured human skinequivalent(Human Skin Equivalent Investigators Group)[J]. Arch Dermatol,1998,134:293–300.
    [169] Eaglstein WH, Alvarez OM, Auletta M, et al. Acute excisional wounds treatedwith a tissue engineered skin (Apligraf)[J]. Dermatol Surg,1999,25:195–201.
    [170] Kim IY, Seo SJ, Moon HS, et a1. Chitosan and its derivatives for tissueengineering applications [J]. Biotechnol Adv,2008,26(1):1-21.
    [171]王旭,王甲汗,吴军,等.复合皮的制作与临床应用[J].中国修复重建外科杂志,1997,11:100-102.
    [172]曹谊林,蔡霞,崔磊,等.自体组织工程化全层皮肤缺损修复实验研究[J].中华外科杂志,2002,20(1):24-27.
    [173]鲁元刚,伍津津,朱堂友,等.复合壳多糖人工皮肤组织学特征研究[J].中国临床康复,2002,6(4):518-519.
    [174] Bottcher-Haberzeth S, Biedermann T, Reichmann E. Tissue engineering ofskin [J]. BURNS,2010,36:450-460.
    [175] Nunery WR (2001) Risk of prion transmission with the use of xenografts andallografts in surgery. Ophthal Plast Reconstr Surg17:389–94.
    [176] Falanga V, Margolis D, Alvarez O, et al. Rapid healing of venous ulcers andlack of clinical rejection with an allogeneic cultured human skin equivalent(Human Skin Equivalent Investigators Group)[J]. Arch Dermatol,1998,134:293–300.
    [177] Lutolf MP, Hubbell JA. Synthetic biomaterials as instructive extracellularmicroenvironments for morphogenesis in tissue engineering [J]. NatBiotechnol,2005,23:47–55.
    [178] Nelson CM, Bissell M. Of extracellular matrix, scaffolds, and signaling: tissuearchitecture regulates development, homeostasis, and cancer [J]. Annu RevCell Dev Biol,2005,22:287–309.
    [179] Putnam AJ, Mooney DJ. Tissue engineering using synthetic extracellularmatrices. Nature Med,1996,2:824–826.
    [180] Heath CA. Cells for tissue engineering [J]. TIBTECH,2000,18:17–19.
    [181] Hubbell JA. Biomaterials in tissue engineering [J]. Bio/technology,1995,13:565-576.
    [182] Marler JJ, Upton J, Langer R, et al. Transplantation of cells in matrices fortissue engineering [J]. Adv Drug Deliv Rev,1998,33:165–182.
    [183] Murphy WL, Mooney DJ. Controlled delivery of inductive proteins, plasmidDNA and cells from tissue engineering matrices [J]. J Periodontal Res,1999,34:413–419.
    [184] Hubbell JA. Bioactive biomaterials [J]. Curr Opin Biotechnol,1999,10:123–129.
    [185] Healy KE. Molecular engineering of materials for bioreactivity [J]. Curr OpinSolid State Mater Sci,1999,4:381–387.
    [186] Sakiyama-Elbert SE, Hubbell JA. Functional biomaterials: design of novelbiomaterials [J]. Annu Rev Mater Res,2001,31:183–201.
    [187] Whitaker MJ, Quirk RA, Howdle SM, et al. Growth factor release from tissueengineering scaffolds [J]. J Pharm Pharmacol,2001,53:1427–1437.
    [188] Richardson TP, Murphy WL, Mooney DJ. Polymeric delivery of proteins andplasmid DNA for tissue engineering and gene therapy [J]. Crit Rev EukaryotGene Expr,2001,11:47–58.
    [189] Babensee JE, McIntire LV, Mikos AG. Growth factor delivery for tissueengineering [J]. Pharm Res,2000,17:497–504.
    [190] Shin H, Jo S, Mikos AG. Modulation of marrow stromal osteoblast adhesionon biomimetic oligo (poly(ethylene glycol) fumarate) hydrogels modifiedwith Arg-Gly-Asp peptides and a poly(ethylene glycol) spacer [J]. J BiomedMater Res,2002,61:169–179.
    [191] Suzuki Y, Tanihara M, Suzuki K, et al. Alginate hydrogel linked withsynthetic oligopeptide derived from BMP-2allows ectopic osteoinduction invivo [J]. J Biomed Mater Res,2000,50:405–409.
    [192]赵长生.生物医用高分子材料[M].北京:化学工业出版社,2009,27-35.
    [193] Ishihara K, Hanyuda H,Nakabayashi N. Synthesis of phospholipi polymershaving a urethane bond in the side chain as coating material on segmentedpolyurethane and their platelet adhesion-resistant properties[J].Biomaterials,1995,16(11):873-879.
    [194] Lewis AL, Hughess PD, Kirkwood LC, et al. Synthesis and characterizationof phosphorylcholine-based polymers useful for coating blood filtrationdevices [J]. Biomaterials,2000,21(18):1847-1859.
    [195] Ishihara K, Fukumoto K, Iwasaki Y, et al. Modification of polysulfone withphospholipid polymer for improvement of the blood compatibility. Part2.Protein adsorption and platelet adhesion [J]. Biomaterials,1999,20(17):1545-1559.
    [196] Lee JH, Kopeckova P,Kopecek J,et al. Suface properities of copolymers ofalkyl methacrylates with methosy (polyethflene oxide) metiiacrylates andtheir application as protein-resistant coatings [J]. Biomaterials,1990,11(7):455-464.
    [197] Quirk RA, Chan WC, Davies MC, et al. Po1y(L-lysine)-GRGDs as abiomimetic surface modifier for Poly(lactic acid)[J]. Biomaterials,2001,22:865-872
    [198] Chen G, Ushida T, Tatsuya T. Hybrid biomaterials for tissueengineering: apreparative method of PLA or PLGA-collagen hybrid sponge [J]. Adv Mater,2000,12:455-457.
    [199] Ma ZW, Gao CY, Gong YH, et al. Paraffin spheres as porogen to fabricatePoly(L-lactic acid) scaffolds with improved cytocompatibility for cartilagetissue engineering [J]. Journal of Biomedical Materials Research B,2003,67B:610-617.
    [200] Zhu HG, Ji J, Lin RY, et al. Surface engineering of Poly (DL-lactic acid) byentrapment of alginate-amino acid derivatives for the promotion ofchondrogenesis [J]. Biomaterials,2002,23:3141-3148.
    [201] Wang Y, Kim JH, Choo KH, et al. Hydrophilic modification of polypropylenemicrofiltration menmbranes by ozone-induced graft polymerization [J].Journal of Membrane Science,2000,169:269-276.
    [202] Dasgupta S. Surface modification of polylefins for hydrophilicity andbondability:Ozonization and grafting hydrophilic monomers on ozonizedpolyolefins [J]. Joumal of Applied Polymer Science,2000,169:269-276.
    [203] Gao CY, Li A, Yi XS, et al. Construction of cell-compatible layer and cultureof human umbilical vaseular endothelial cells on porous polystyrenemembranes [J]. Joumal of Applied Polylmer Science,2001,81:3523-3529.
    [204] Cai KY, Yao KD, Lin SB, et al. Poly (D,L-lactic acid) surface modified bysilk fibroin:effects on the culture of osteoblast in vitro [J]. Biomaterials,2002,23:1153-1160.
    [205] Zhu YB, Gao CY, Liu XY, et al. Surface modification polycaprolactonemembrane via aminolysis and biomacromolecule immobilization forpromoting cytocompatibility of human endothelial cells [J].Biomacromolecules,2002,3:1312-1319.
    [206] Chu PK, Chen JY, Wang LP, et al. Plasma-surface modification ofbiomaterials [J]. Mater.Sci.Eng.R.,2002,36:143-206.
    [207]赵剑豪,屠美,田冶,等.等离子体处理偶联胶原提高聚乳酸材料的细胞相容性[J].高分子材料科学与工程,2008,24(7):140-143.
    [208] Zhu AP, Chen T. Blood compatibility of surface-engineered poly(ethyleneterephtha-late)via O-carboxymethylchitosan [J]. Colloids and Surfaces B:Bio-interfaces,2006,50(2):120-125.
    [209]郑玉峰,李莉.生物医用材料学[M].哈尔滨:哈尔滨工业大学出版社,2006,8:349-352.
    [210]石淑先.生物材料制备与加工[M].北京:化学工业出版社,2008,9:420-436.
    [211] Colwell JM, Wentrup BE, Bell JM, et al. A study of the chemical and physicaleffects of ion implantation of micr-porous and nonporous PTFE [J]. Surfaceand Coatings Technology,2003,168(2/3):216-222.
    [212] Sato H, Tsuji H, Ikemoto N, et al. Enhanced growth of human vaseularendothelial cells on negative ion(Ag)一imLanted hydrophobic surfaces [J]. JBiomed Mater Res,1999,44:22-30.
    [213] Khang G, Jeon JH, Lee JW, et al. Platelet and cell interactions on goldsputter-deposited polymeric surfaces [J]. Bio-medical Materials andEngineering,1998,8:299-309.
    [214] Blodgett KB. Films built by depositing successive monomolecular layers on asolid suface [J]. Journal of the American Chemical Society, l935,57(6):1007-1022.
    [215] Lucy N, Jacob S. A new approach to construction of artificial monolayerassemb1ies [J]. J. Am.Chem.Soc.,1983,105(3):674-676.
    [216] Deeher G, Hong JD, Schmitt J. Buildup of ultrathin multilayer films by aself-assemnly proeess: Consecutively alternating adsorption of anionic andcationic polyelectrolytes on charged surface [J]. Thin Solid Films,1992,210:831-835.
    [217] Schlenoff J. Redox-active polyelectrolyte multilayers [J]. Adv.Mater,1998,10:347-351.
    [218] Mendelson J, Barrett C, Chan V, et al. Fabrication of microporous thin filmsfrom polyelectrolyte multilayers [J]. Langmuir,2000,16:5017-5023.
    [219] Sullivan DM, Bruening ML. Ultrathin ion-selective polyimide membranesprepared from layered polyelectrolytes [J]. J.Am.Chem.Soc,2001,123:11805-11806.
    [220] Chung AJ, Rubner MF. Methods of loading and releasing low molecularweight cationic molecules in weak polyelectrolyte multilayer films [J].Langmuir,2002,18(4):1176-1183.
    [221] Vazquez E, Dewitt DM, Hammond PT, et al. Construction ofhydrolytically-degradable thin films via layer-by-layer deposition ofdegradable polyelcetrolytes [J]. J Am Chem Soc,2002,124(47):13992-13993.
    [222] Thierry B, Winnik F, Merhi Y, et al. Nanocoatings onto Arteries viaLayer-by-Layer Deposition:Towards the in-vivo Repair of Damaged BloodVessels [J]. Joumal of the American Chemieal Society,2003,125(25):7494-7495.
    [223] Donald LE, Curtis BH, Jeffrey AH. Thin Polymer Layers Formed byPolyelectrolyte Multilayer Techniques on Biological Sufaces [J]. Langmuir,1999,15(16):5355-5362.
    [224] Sung YY, Jonas DM, Michael FR. New Class of Ultrathin, HighlyCell-Adhesion-Resistant Polyelectrolyte Multilayers with MicropatteringCapabilities [J]. Biomacromolecules,2003,4(4):987-994.
    [225] Ji J, Tan QG, Shen JC, et al. Fabrication of alternating polycation and albuminmultilayer coating onto stainless steel by electrostatic layer-by-layeradsorption [J]. Colloids and Surfaces B: Biointerfaces,2004,34:185-190.
    [226] Lvov Y, Decher G, Sukhorukov G. Assembly of thin films by means ofsuccessive deposition of alternate layers of DNA and poly(allylamine)[J].Macromolecules,1993,26(20):5396-5399.
    [227] Richert L, Lavalle P, Picart C, et al. Layer by Layer Buildup ofPolysaccharide Films: Physical Chemistry and Cellular Adhesion Aspects [J].Langmuir,2004,20:448-458.
    [228] Brynda E, Houska M, Jirouskova M, et al. Albumin and heparin multilayercoatings for blood-contacting medcial devices [J]. Journal of BiomedicalMaterials Research,2000,51(2):249-257.
    [229] Guire PE. Methods in Enzymology: USA,4722906[P/OL].1982-09-29[1988-02-02].
    [230] Friedberg. Affinity Chromatography and Insoluble Enzymes [J].Chromatogr Rev,1971,14:121-131.
    [231] S Aiba, N Minoura, K Taguchi, Y Fujiwara. Covalent immobilization ofchitosan derivatives onto polymeric film surfaces with the use of aphotosensitive hetero-bifunctional crosslinking reagent [J]. Biomatenals,1987,8(6):481.
    [232] T. Matsuda, K. Inoue, T. Sugawara. Development of micropatterningtechnology for cultured cells [J]. Trans Am Soc Artif Intern Organs,1990,36:M559–M563.
    [233] Clapper DL. Cardiovascular Science and Technology: Basic and Applie [C],Louis-ville,kY.Dec.1-3,1990.
    [234] Colly LE, Guire PE. Light activated polymers for flexible surfacemodification [C]. Prceeding of the Medical Design&ManufacturingConference, June3,1992, NewYork,USA.
    [235] Amos RA, Guire RE. Surface modification of polymers by photochemicalimmobilization-a general method [C]. The17thAnnual meeting of the societyfor Biomaterials May l-5,1991.
    [236] Gyorgy Dorman, GlemD. Prestwich benzophenone photophores inbiochemistry [J]. Biochemistry,1994,(33)9:5661-5673.
    [237] Y.Uyama, H.Tdokoro, Y.Ikada. Surface lubrication of polymer films byphotoinduced graft polyrmerization [J]. J Appl Sci, l990,(39):489-498.
    [238] Matsuda Takhisa.Photochemically-driven surface process technologies forbiocompatible design off fabricated cardiovascular devices [C]. The23rdannual meeting of the society for biomaterials. April30-May4,1997.
    [239] Swanson MJ, Dunkirk SG, Pietig JA, et al. Use photochemistry to modifymembranes: the technique converts hydrophobic surfaces to hydrophilic,making them more useful for certain biochemical applications [J]. Chemtech1992,10:624.
    [240] Arnos RA, Guire PE, Trans Soc. For Bionmat.1991,XIV:175.
    [241] Duran LW, Pieting JA, Driermeyer JE, et al. The19thannual Meeting of theSociety for Biomaterials, Apirl28-May l,1993,Birmingham AL, USA.
    [242]王文文,陶慧敏,李志斌,等.共固定化TNF-α/IFN-γ对宫颈癌HeLa细胞的长效抑制作用[J].离子交换与吸附,20099,25(2):130-136.
    [243] Yoshihiro Ito, GuoPing Chen, Yukio Imamsh, et al. Photoimmobilization ofSulfated Hyaluronic Acid for Antithrombogenicity.Biotechnol.Prog.1997,8(5):730-734.
    [244] Matsuda T, Sugawara T. Photochemical protein fixation on polymer surfacesvia derivatized phenylazido group [J]. Langmuir,1995,11:2272-2276.
    [245] Tae Il Sona, Makoto Sakuragi, Sawa Takahashi, et al. Visible light-inducedcrosslinkable gelatin (Visible light-induced crosslinkable gelatin)[J]. ActaBiomaterialia,2010,6(10):4005-4010.
    [246] Rodolfo E. Gámez Sazo, Katsumi Maenaka, Weiyong Gu, et al. Fabricationof growth factor and extracellular matrix-loaded, gelatin-based scaffolds andtheir biocompatibility with Schwann cells and dorsal root ganglia.Biomaterials,2012,33(11):8529-8539.
    [247] Aiping Zhu, Feng Zhao, Teng Ma. Photo-initiated grafting of gelatin/N-maleicacyl-chitosan to enhance endothelial cell adhesion, proliferation and functionon PLA surface [J]. Acta Biomaterialia,2009,5(6):2033-2044.
    [248]崔巍巍.载银PLGA抗菌敷料的制备及毒性评价[D].吉林:吉林大学公共卫生学院,2010.
    [249]徐冰函.聚乙烯醇水凝胶制备及生物评价[D].吉林:吉林大学公共卫生学院,2011.
    [250] Xin XJ, Hussain M, Mao JJ. Self replication of human mesenchymal stemcells and continuing differentiation of derived chondrogenic and osteogeniclineages in electrospun PLGA nanofiber scaffold [J]. Biomaterials,2007,28(2):316-325.
    [251] L. Lu, G.N. Stamatas, A.G. Mikos. Controlled release of transforming growthfactor beta1from biodegradable polymer microparticles [J].Journal ofBiomedical Materials Research,2000,50:440-451.
    [252] Katti DS, Laurencin CT. Synthetic biomedical polymers for tissue engineeringand drug delivery. In: Advani S, Shonaike GO, editors. Advanced polymericmaterials: Structure property relationship [M].Boca Raton, FL: CRC Press,2003:479-526.
    [253] E. Pulieri, V. Chiono, G. Ciardelli, G. Vozzi, et al. Chitosan/gelatin blends forbiomedical applications [J]. Journal of Biomedical Materials Research Part A,2008,86(2):311-322.
    [254] Guillem Rocasalbas, Antonio Francesko, Sonia Touri o, et al. Laccaseassisted formation of bioactive chitosan/gelatin hydrogel stabilized with plantpolyphenols [J]. Carbohydrate Polymers,2013,92(2):989-996.
    [255] Kezban Ulubayram, A.Nur Cakar, Petek Korkusuz, et al. EGF containinggelatin-based wound dressings [J]. Biomaterials,2001,22(11):1345-1356.
    [256] Masaya Ohsaki, Hiroyuki Tsutsumi, Takuji Kumagai, et al. The relevance ofTH1and TH2cells in immediate and nonimmediate reactions togelatin-containing vaccine [J]. Journal of Allergy and Clinical Immunology,1999,103(2):276-281.
    [257] Jacqueline D Kosmala, David B Henthorn, Lisa Brannon-Peppas, et al.Preparation of interpenetrating networks of gelatin and dextran as degradablebiomaterials [J]. Biomaterials,2000,21(10):2019-2023.

© 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