BMP-2和BMSCs结合n-HA/n-LDIG复合材料修复兔桡骨缺损的实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
第一章兔骨髓间充质干细胞的分离、培养、鉴定及诱导分化成骨细胞
     目的:探讨兔BMSCs的分离、培养、鉴定及向成骨细胞诱导分化的方法,探讨BMSCs在体外增殖,定向分化能力。
     方法:取兔股骨的骨髓冲洗液,采用Percoll分离液密度梯度离心法结合贴壁筛选法分离、培养、纯化兔BMSCs,光镜下观察传代细胞生长状况和形态变化;MTT法检测细胞增殖情况,绘制生长动力学曲线;HE染色形态观察和流式细胞仪抗体鉴定兔BMSCs表面标记物;体外诱导P3BMSCs向成骨细胞分化,对诱导分化为骨细胞进行Gomori钙钴法ALP染色和茜素红钙矿化结节染色鉴定向成骨分化的能力。
     结果:所分离培养的细胞形态均一,沿胞体长轴生长,呈旋涡状、网状、梭状、放射状,形成较典型的BMSCs。P3BMSCs的生长曲线大致呈S型,第7-11天细胞处于高速增长期,细胞数量迅速增加;HE染色BMSCs形态呈长梭形或长三角形,细胞核浆比例大,核蓝染,胞浆丰富红染。流式检测细胞表面标记物显示BMSCsCD44表达百分率为98.63%,呈阳性,而CD45表达呈阴性。P3BMSCs在体外经成骨诱导3周后,Gomori钙钴法染色ALP呈阳性,胞浆显示有灰黑色颗粒或棕黑色或黑色沉淀。茜素红染色呈阳性,细胞间出现致密的圆形或者卵圆形矿化结节。
     结论:应用Percoll分离液密度梯度离心法和贴壁筛选法结合能分离、培养出兔BMSCs,是一种比较理想获取兔BMSCs的方法;兔P3BMSCs纯度较高和良好的增殖能力,具有向成骨细胞定向分化的生物学特性,是组织工程理想的种子细胞,也为后续进行骨缺损修复实验奠定了坚实的基础。
     第二章n-HA/n-LDIG复合材料的制备及与BMSCs体外复合培养的细胞生物学评价
     目的:以赖氨酸盐,甘油及n-HA为主要原料,制备出一种新型n-HA/n-LDIG复合材料,分析其与BMSCs体外复合培养的生物相容性,探讨其作为组织工程支架的可行性。
     方法:选择赖氨酸盐和甘油为原材料,通过有机合成、聚合反应、纳米化、超临界抗溶剂结晶、超声分散等技术制备出n-HA/n-LDIG复合材料。电镜观察其结构及计算孔隙率,细胞计数法检测细胞对复合材料的黏附能力,MTT法检测复合材料对细胞增殖的影响,ALP活性和钙含量测定观察复合材料对细胞分化的影响。
     结果:n-HA/n-LDIG复合材料肉眼为白色散在,为比较均匀的粉状固体。扫描电镜可见类似天然松质骨的三维孔洞网络结构,透射电镜可见排列不规则的晶须和空穴。共培养从2h至10h,随时间细胞的黏附率有所增加,其中6h时细胞黏附率明显高于4h时,8h时明显高于6h时,其差异均有统计学意义(P<0.05)。MTT实验结果显示共培养第2天到第16天,细胞在复合材料上数量随时间而增加,特别是从第6天开始复合材料上的细胞数量迅速增加,细胞处于对数生长期,12天后过渡到生长平台期,增殖缓慢。从第4天开始,黏附细胞的ALP相对活性和钙相对含量迅速上升;第8天时ALP相对活性和钙相对含量明显高于第4天时,第12天明显高于第8天时,其差异均有统计学意义(P<0.05);12天后ALP相对活性和钙相对含量增加不明显,差异无统计学意义。
     结论:制备的n-HA/n-LDIG复合材料具备适宜的孔隙率和孔径,组成性质相似于天然骨结构,具有良好的生物学特性,是一种新型的纳米骨修复材料。n-HA/n-LDIG复合材料具备良好的细胞相容性,能为成骨细胞在其表面生长、增殖、分泌基质提供良好的微环境,是一种良好的骨修复替代材料,为其作为骨的快速修复和骨组织工程支架材料奠定了基础。
     第三章BMP-2和BMSCs结合n-HA/n-LDIG复合材料修复兔桡骨缺损的实验研究
     目的:构建兔桡骨缺损模型,探讨BMP-2和BMSCs结合n-HA/n-LDIG复合材料修复兔桡骨缺损的效果。
     方法:取新西兰大白兔构建兔桡骨缺损模型,实验组植入BMP-2和BMSCs结合n-HA/n-LDIG复合材料,对照组植入BMSCs结合n-HA/n-LDIG复合材料,空白组不植入任何材料。分别于2、4、8、12周随机处死家兔,进行动物大体观察,X线检查和Lane-Sandhu放射学评分观察骨缺损处愈合情况,通过组织学检查和Lane-Sandhu组织学评分观察骨缺损处的修复情况。
     结果:所有实验动物桡骨骨缺损模型构建均成功,未见皮下和肌肉组织充血、水肿、渗出及感染等现象。从大体观察,X线检查和组织学检查等发现随着时间的延长,实验组修复效果优于对照组,对照组优于空白组。实验组术后3、6、9周新生骨所占骨缺损面积百分比,Lane-Sandhu放射学评分,新生骨所占骨缺损面积百分比及Lane-Sandhu组织学评分均明显高于对照组,对照组均明显高于空白组,其差异均具有统计学意义(P<0.05)。
     结论:BMP-2明显够促进早期成骨,缩短成骨时间,加速骨缺损区骨组织的再生和钙化;BMP-2和BMSCs结合n-HA/n-LDIG复合材料具有良好的骨缺损修复效果,能明显够促进早期成骨,缩短成骨时间,加速骨缺损区骨组织的再生和钙化,能为临床应用提供了理论基础和新思路,具有良好的临床应用前景。
Chapter1Separation, Culture, Identification of Rabbit Bone Marrow Mesenchymal Stem Cells and Their Induced Differentiation to Osteoblasts
     Objective:To explore the separation, culture, identification of rabbit bone marrow mesenchymal stem cells (BMSCs) and their induced differentiation to osteoblasts, as well as the capacity of BMSCs in proliferation in vitro and directed differentiation.
     Method:The bone marrow washing fluid of rabbit femur was taken for separation, culture, purified rabbit of BMSCs by Percoll separation medium density gradient centrifugation and adherence screening method, the growth conditions and morphological changes of passaged cells were observed under light microscope; the cell proliferation was determined by MTT assay so as to fit growth kinetic curves; the rabbit BMSCs surface markers were observed by HE staining morphology and identified by flow cytometry antibodies; the P3BMSCs were induced in vitro to differentiate into the osteoblasts, the induced BMSCs were stained by Gomori Gomori ALP staining and alizarin Red calcium mineralized nodules to identify their capacity to differentiate into osteoblasts.
     Results:The cells for separated culture were highly homogeneous, and grew along the long axis of cell body, presented swirly, mesh, fusiform, radial, and formed typical BMSCs. The growth curve of P3BMSCs was roughly S-shaped, their7~11d cells were in a period of rapid growth, the number of cells here rapidly increased; the HE staining BMSCs morphology presented long fusiform or long triangle, the nucleus cytoplasm ratio was high, the nuclear was blue-stained, the cytoplasm was abundantly red-stained. The flow cytometry detection for the cell surface markers showed the expression percentage of the BMSCs CD44was98.63%, positive, while the expression of CD45presented negative. At3weeks after the P3BMSCs were induced to osteoblasts in vitro, the Gomori calcium cobalt staining ALP was positive, the cytoplasm showed gray and black particles or brown-black or black precipitates. Alizarin Red staining was positive, the intercellular space appeared round or oval dense mineralized nodules.
     Conclusion:The rabbit BMSCs can be separated and cultured through the combination of Percoll separated medium density gradient centrifugation and adherence screening method which is an ideal method to obtain rabbit BMSCs; rabbit P3BMSCs have good purity, good proliferative capacity, and biological characteristics of directed differentiation to osteoblasts, which is a kind of ideal seed cells for tissue engineering and also lays a solid foundation for follow-up bone defect repair experiments.
     Chapter2Rreparation of n-HA/n-LDIG composites and biology assessment for the cells compoundly cultured from bmscs in vitro
     Objective:The lysine salt, glycerine and n-HA were selected as the main raw materials to prepare a new n-HA/n-LDIG composite, analyzing its compatibility to the cells compoundly cultured from BMSCs in vitro so as to explore its feasibility to be a tissue engineering scaffold.
     Methods:The lysine salt and glycerol were selected as raw materials, through organic synthesis, polymerization, nanotechnology, supercritical anti-solvent crystallization, ultrasonic dispersion technologies, they were processed to prepare a n-HA/n-LDIG composite. By electron microscopy, its structure and porosity was observed and calculated, then the cell adhesion of it was determined by cell counting method, and its influence on cell proliferation was determined by MTT assay, next its influence on cell differentiation was determined by ALP activity and calcium content determination.
     Results:The n-HA/n-LDIG composite visually likes scatteredly white, relatively uniform powdery solid. The3D porous network structure similar to natural cancellous bone was observed under the scanning of electron microscopy, and its irregular arrangement of whiskers and pores could be found under transmission electron microscopy. The cells were cultured within2h to10h, with the time increased, the cell adhesion also increased, and the cell adhesion rate at6h was significantly higher than that at4h, the cell adhesion rate at8h was significantly higher than that at6h, their differences were statistically significant (P<0.05). MTT assay results showed:from2d to16d of culture, with the time increased, the cell counts on composite also increased, especially rapidly increased from6d (the cells were in the exponential growth phase), on12d, the cells developed to a growth plateau with slow proliferation. From4d, the ALP relative activity and calcium relative content quickly raised; the ALP relative activity and calcium relative content on8d were significantly higher than those on4d, and the ALP relative activity and calcium relative content on12d were significantly higher than those on8d, their differences were statistically significant (P<0.05); from12d, the growth rate of ALP relative activity and calcium relative content slowed down, their difference was not statistically significant (P>0.05).
     Conclusions:The prepared n-HA/n-LDIG composite has suitable porosity and pore size, and its composition properties are similar to those of natural bone structures, having good biological characteristics, so it is a new nano-bone repair material. The n-HA/n-LDIG composite have good cell compatibility and can act as microenvironment for the growth, proliferation, matrix secretion of cells on its surface. As a premium bone repair alternative material, it laid a foundation for rapid bone repair and bone tissue engineering scaffold material development.
     Chapter3Experimental study on repairmen of rabbit radial bone defects using BMP-2and BMSCs combined with n-HA/n-LDIG composite material
     Objective:To build the rabbit radial bone defect model, and explore the repair effects of rabbit radial bone defects using BMP-2and BMSCscombined with n-HA/n-LDIG composite material.
     Method:The New Zealand white rabbits were used to build bone defects models, the experiment group was implanted BMP-2and BMSCs combined with n-HA/n-LDIG composite material, and the control group was implanted the BMSCs combined with n-HA/n-LDIG composite material, and the blank group was not implanted. The rabbits were randomly sacrificed after2,4,8and12weeks and taken the general observation, X-ray examination and Lane-Sandhu scoring to observe the healing of the bone defect, and through the histological examination and Lane-Sandhu histological scoring to observe the repair of the bone defect.
     Results:All experimental animal radius bone defect models were successfully built, subcutaneous and muscle tissue congestion, edema, exudate and infection were not observed. From the general observation, X-ray examination and histological examination, we can found that, with the time going, the repair effects of the experiment group were better than that of the control group, and the repair effects of the control group were better than that of the blank group. The area percentage of newborn bone of bone defect, Lane-Sandhu radiological scores and the Lane-Sandhu histological scores after3,6and9weeks of the experiment group were significantly higher than of the control group, and the control group were better than the blank group, and the differences were statistically significant (P<0.05).
     Conclusion:The BMP-2can significantly promote the early osteogenesis, shorten the osteogenesis time and accelerate the regeneration and calcification of the bone tissue at bone defect area; the BMP-2and BMSCs combined with n-HA/n-LDIG composite material has good bone defect repair effects, can significantly promote the early osteogenesis, shorten the osteogenesis time and accelerate the regeneration and calcification of the bone tissue at bone defects area, provides a theoretical basis and new idea for clinical application, and has good prospects for clinical application.
引文
[1]Ilmer M, Karow M, Geissler C, et al. Human osteoblast-derived factors induce early osteogenic markers in human mesenchymal stem cells[J]. Tissue Eng Part A,2009,15(9):2397-409.
    [2]Zhu H, Jiang XX, Wu Y, et al. Identification of mesenchymal stem cells derived from rheumatoid arthritis synovial fluid and their regulatory effect on osteoblast formation[J]. Zhongguo Shi Yan Xue Ye Xue Za Zhi,2009,17(4): 977-80.
    [3]Reinshagen H, Auw-Haedrich C, Sorg RV, et al. Corneal surface reconstruction using adult mesenchymal stem cells in experimental limbal stem cell deficiency in rabbits[J]. Acta Ophthalmol,2011,89(8):741-8.
    [4]Feng G, Zhao X, Liu H, et al. Transplantation of mesenchymal stem cells and nucleus pulposus cells in a degenerative disc model in rabbits:a comparison of 2 cell types as potential candidates for disc regeneration[J]. J Neurosurg Spine, 2011,14(3):322-9.
    [5]Jiang SJ, He XS, Ni YD, et al. Proliferation of the mesenchymal stem cells in a delayed fat flap:an experimental study in rabbits[J]. Zhonghua Zheng Xing Wai Ke Za Zhi,2009,25(4):287-9.
    [6]Du XF, Kwon SK, Song JJ, et al. Tracheal reconstruction by mesenchymal stem cells with small intestine submucosa in rabbits[J]. Int J Pediatr Otorhinolaryngol,2012,76(3):345-51.
    [7]Lee JY, Choi MH, Shin EY, et al. Autologous mesenchymal stem cells loaded in Gelfoam((R)) for structural bone allograft healing in rabbits[J]. Cell Tissue Bank,2011,12(4):299-309.
    [8]Bo B, Wang CYGuo XM. Repair of cranial defects with bone marrow derived mesenchymal stem cells and beta-TCP scaffold in rabbits[J]. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi,2003,17(4):335-8.
    [9]Udehiya RK, Amarpal, Aithal HP, et al. Comparison of autogenic and allogenic bone marrow derived mesenchymal stem cells for repair of segmental bone defects in rabbits[J]. Res Vet Sci,2013.
    [10]Nather A, David V, Teng JW, et al. Effect of autologous mesenchymal stem cells on biological healing of allografts in critical-sized tibial defects simulated in adult rabbits[J]. Ann Acad Med Singapore,2010,39(8):599-606.
    [11]Ruiz-Iban MA, Diaz-Heredia J, Garcia-Gomez I, et al. The effect of the addition of adipose-derived mesenchymal stem cells to a meniscal repair in the avascular zone:an experimental study in rabbits[J]. Arthroscopy,2011,27(12): 1688-96.
    [12]Dashtdar H, Rothan HA, Tay T, et al. A preliminary study comparing the use of allogenic chondrogenic pre-differentiated and undifferentiated mesenchymal stem cells for the repair of full thickness articular cartilage defects in rabbits[J]. J Orthop Res,2011,29(9):1336-42.
    [13]Lin CY, Chang YH, Lin KJ, et al. The healing of critical-sized femoral segmental bone defects in rabbits using baculovirus-engineered mesenchymal stem cells[J]. Biomaterials,2010,31(12):3222-30.
    [14]Kim SS, Song CK, Shon SK, et al. Effects of human amniotic membrane grafts combined with marrow mesenchymal stem cells on healing of full-thickness skin defects in rabbits[J]. Cell Tissue Res,2009,336(1):59-66.
    [15]Ohya M, Yamada Y, Ozawa R, et al. Sinus floor elevation applied tissue-engineered bone. Comparative study between mesenchymal stem cells/platelet-rich plasma (PRP) and autogenous bone with PRP complexes in rabbits[J]. Clin Oral Implants Res,2005,16(5):622-9.
    [16]Song YS, Ku JH, Song ES, et al. Magnetic resonance evaluation of human mesenchymal stem cells in corpus cavernosa of rats and rabbits[J]. Asian J Androl,2007,9(3):361-7.
    [17]Fujita H, Kurokawa K, Ogino T, et al. Effect of risedronate on osteoblast differentiation, expression of receptor activator of NF-kappaB ligand and apoptosis in mesenchymal stem cells[J]. Basic Clin Pharmacol Toxicol,2011, 109(2):78-84.
    [18]Abdallah BM, Haack-Sorensen M, Fink T, et al. Inhibition of osteoblast differentiation but not adipocyte differentiation of mesenchymal stem cells by sera obtained from aged females[J]. Bone,2006,39(1):181-8.
    [19]Sowa H, Kaji H, Canaff L, et al. Inactivation of menin, the product of the multiple endocrine neoplasia type 1 gene, inhibits the commitment of multipotential mesenchymal stem cells into the osteoblast lineage[J]. J Biol Chem,2003,278(23):21058-69.
    [20]Kulterer B, Friedl G, Jandrositz A, et al. Gene expression profiling of human mesenchymal stem cells derived from bone marrow during expansion and osteoblast differentiation[J]. BMC Genomics,2007,8(70).
    [21]Munisso MC, Kang JH, Tsurufuji M, et al. Cilomilast enhances osteoblast differentiation of mesenchymal stem cells and bone formation induced by bone morphogenetic protein 2[J]. Biochimie,2012,94(11):2360-5.
    [22]Wu X, Estwick SA, Chen S, et al. Neurofibromin plays a critical role in modulating osteoblast differentiation of mesenchymal stem/progenitor cells[J]. Hum Mol Genet,2006,15(19):2837-45.
    [23]Sun J, Zhou H, Deng Y, et al. Conditioned medium from bone marrow mesenchymal stem cells transiently retards osteoblast differentiation by downregulating runx2[J]. Cells Tissues Organs,2012,196(6):510-22.
    [24]Zhao JW, Gao ZL, Mei H, et al. Differentiation of human mesenchymal stem cells:the potential mechanism for estrogen-induced preferential osteoblast versus adipocyte differentiation[J]. Am J Med Sci,2011,341(6):460-8.
    [25]Sethi S, Radio NM, Kotlarczyk MP, et al. Determination of the minimal melatonin exposure required to induce osteoblast differentiation from human mesenchymal stem cells and these effects on downstream signaling pathways[J]. J Pineal Res,2010,49(3):222-38.
    [26]Plaisant M, Fontaine C, Cousin W, et al. Activation of hedgehog signaling inhibits osteoblast differentiation of human mesenchymal stem cells[J]. Stem Cells,2009,27(3):703-13.
    [27]Dahl M, Syberg S, Jorgensen NR, et al. Adipose derived mesenchymal stem cells-Their osteogenicity and osteoblast in vitro mineralization on titanium granule carriers[J]. J Craniomaxillofac Surg,2013.
    [28]Zhu F, Guo GH, Huang S, et al. The effect of bone marrow mesenchymal stem cells engraftment on lung tissue at early stage of smoke inhalation injury in rabbits[J]. Zhongguo Wei Zhong Bing Ji Jiu Yi Xue,2011,23(1):18-20.
    [29]Zhao M, Zhou J, Li X, et al. Repair of bone defect with vascularized tissue engineered bone graft seeded with mesenchymal stem cells in rabbits [J]. Microsurgery,2011,31(2):130-7.
    [30]Kim BS, Kim JS, Sung HM, et al. Cellular attachment and osteoblast differentiation of mesenchymal stem cells on natural cuttlefish bone[J]. J Biomed Mater Res A,2012,100(7):1673-9.
    [31]Nicolaidou V, Wong MM, Redpath AN, et al. Monocytes induce STAT3 activation in human mesenchymal stem cells to promote osteoblast formation[J]. PLoS One,2012,7(7):e39871.
    [32]Backesjo CM, Li Y, Lindgren U, et al. Activation of Sirtl decreases adipocyte formation during osteoblast differentiation of mesenchymal stem cells[J]. Cells Tissues Organs,2009,189(1-4):93-7.
    [33]Mathews S, Mathew SA, Gupta PK, et al. Glycosaminoglycans enhance osteoblast differentiation of bone marrow derived human mesenchymal stem cells[J]. J Tissue Eng Regen Med,2012.
    [34]Lim TY, Wang W, Shi Z, et al. Human bone marrow-derived mesenchymal stem cells and osteoblast differentiation on titanium with surface-grafted chitosan and immobilized bone morphogenetic protein-2[J]. J Mater Sci Mater Med,2009,20(1):1-10.
    [35]Laurencin CT, Khan Y, Kofron M, et al. The ABJS Nicolas Andry Award: Tissue engineering of bone and ligament:a 15-year perspective[J]. Clin Orthop Relat Res,2006,447(221-36.
    [36]Meyer U, Joos UWiesmann HP. Biological and biophysical principles in extracorporal bone tissue engineering. Part I[J]. Int J Oral Maxillofac Surg, 2004,33(4):325-32.
    [37]Wiesmann HP, Joos UMeyer U. Biological and biophysical principles in extracorporal bone tissue engineering. Part II[J]. Int J Oral Maxillofac Surg, 2004,33(6):523-30.
    [38]Liebschner MA. Biomechanical considerations of animal models used in tissue engineering of bone[J]. Biomaterials,2004,25(9):1697-714.
    [39]Jose MV, Thomas V, Xu Y, et al. Aligned bioactive multi-component nanofibrous nanocomposite scaffolds for bone tissue engineering[J]. Macromol Biosci,2010,10(4):433-44.
    [40]Jose MV, Thomas V, Johnson KT, et al. Aligned PLGA/HA nanofibrous nanocomposite scaffolds for bone tissue engineering[J]. Acta Biomater,2009, 5(1):305-15.
    [41]Delabarde C, Plummer CJ, Bourban PE, et al. Biodegradable polylactide/hydroxyapatite nanocomposite foam scaffolds for bone tissue engineering applications[J]. J Mater Sci Mater Med,2012,23(6):1371-85.
    [42]Bhattacharyya S, Kumbar SG, Khan YM, et al. Biodegradable polyphosphazene-nanohydroxyapatite composite nanofibers:scaffolds for bone tissue engineering[J]. J Biomed Nanotechnol,2009,5(1):69-75.
    [43]Duan B, Wang M, Zhou WY, et al. Three-dimensional nanocomposite scaffolds fabricated via selective laser sintering for bone tissue engineering[J]. Acta Biomater,2010,6(12):4495-505.
    [44]Jayakumar R, Chennazhi KP, Srinivasan S, et al. Chitin scaffolds in tissue engineering[J].Int J Mol Sci,2011,12(3):1876-87.
    [45]Nowiek JS, Powell NA, Nguyen TM, et al. An improved method for the synthesis of enantimerically pure amino acid ester isocyanates[J].J OrgChem, 1992,57:7364-736.
    [46]Zhang JY, Beckman EJ, Piesco NP, et al. A new peptide-based urethane polymer:synthesis, biodegradation, and potential to support cell growth in vitro[J]. Biomaterials,2000,21(12):1247-58.
    [47]李坚.高分子纳米材料的研制及用于骨组织工程的初步研究[D].长沙:中南大学,2006.
    [48]Kruyt MC, van Gaalen SM, Oner FC, et al. Bone tissue engineering and spinal fusion:the potential of hybrid constructs by combining osteoprogenitor cells and scaffolds[J]. Biomaterials,2004,25(9):1463-73.
    [49]Cowan CM, Soo C, Ting K, et al. Evolving concepts in bone tissue engineering[J]. Curr Top Dev Biol,2005,66(239-85.
    [50]Cui H, Liu Y, Deng M, et al. Synthesis of biodegradable and electroactive tetraaniline grafted poly(ester amide) copolymers for bone tissue engineering[J]. Biomacromolecules,2012,13(9):2881-9.
    [51]De Oliveira AA, de Carvalho SM, Leite Mde F, et al. Development of biodegradable polyurethane and bioactive glass nanoparticles scaffolds for bone tissue engineering applications[J]. J Biomed Mater Res B Appl Biomater, 2012,100(5):1387-96.
    [52]Duan BWang M. Customized Ca-P/PHBV nanocomposite scaffolds for bone tissue engineering:design, fabrication, surface modification and sustained release of growth factor[J]. J R Soc Interface,2010,7 Suppl 5(S615-29.
    [53]El-Ghannam A, Ning CQMehta J. Cyclosilicate nanocomposite:a novel resorbable bioactive tissue engineering scaffold for BMP and bone-marrow cell delivery[J]. J Biomed Mater Res A,2004,71(3):377-90.
    [54]Fernandez JM, Molinuevo MS, Cortizo MS, et al. Development of an osteoconductive PCL-PDIPF-hydroxyapatite composite scaffold for bone tissue engineering[J]. J Tissue Eng Regen Med,2011,5(6):e126-35.
    [55]Fricain JC, Schlaubitz S, Le Visage C, et al. A nano-hydroxyapatite--pullulan/dextran polysaccharide composite macroporous material for bone tissue engineering[J]. Biomaterials,2013,34(12):2947-59.
    [56]Hankemeier S, van Griensven M, Ezechieli M, et al. Tissue engineering of tendons and ligaments by human bone marrow stromal cells in a liquid fibrin matrix in immunodeficient rats:results of a histologic study[J]. Arch Orthop Trauma Surg,2007,127(9):815-21.
    [57]Kheir E, Stapleton T, Shaw D, et al. Development and characterization of an acellular porcine cartilage bone matrix for use in tissue engineering[J]. J Biomed Mater Res A,2011,99(2):283-94.
    [58]Kim HJ, Kim UJ, Vunjak-Novakovic G, et al. Influence of macroporous protein scaffolds on bone tissue engineering from bone marrow stem cells[J]. Biomaterials,2005,26(21):4442-52.
    [59]Lacroix D, Chateau A, Ginebra MP, et al. Micro-finite element models of bone tissue-engineering scaffolds[J]. Biomaterials,2006,27(30):5326-34.
    [60]Barbanti SH, Santos AR, Jr., Zavaglia CA, et al. Poly(epsilon-caprolactone) and poly(D,L-lactic acid-co-glycolic acid) scaffolds used in bone tissue engineering prepared by melt compression-particulate leaching method[J]. J Mater Sci Mater Med,2011,22(10):2377-85.
    [61]Boyan BD, Ranly DMSchwartz Z. Use of growth factors to modify osteoinductivity of demineralized bone allografts:lessons for tissue engineering of bone[J]. Dent Clin North Am,2006,50(2):217-28, viii.
    [62]Chung EJ, Kodali P, Laskin W, et al. Long-term in vivo response to citric acid-based nanocomposites for orthopaedic tissue engineering[J]. J Mater Sci Mater Med,2011,22(9):2131-8.
    [63]Egli RJLuginbuehl R. Tissue engineering-nanomaterials in the musculoskeletal system[J]. Swiss Med Wkly,2012,142(w13647.
    [64]Fu SZ, Wang XH, Guo G, et al. Preparation and properties of nano-hydroxyapatite/PCL-PEG-PCL composite membranes for tissue engineering applications [J]. J Biomed Mater Res B Appl Biomater,2011, 97(1):74-83.
    [65]Garcia-Garcia JM, Garrido L, Quijada-Garrido I, et al. Novel poly(hydroxyalkanoates)-based composites containing Bioglass(R) and calcium sulfate for bone tissue engineering[J]. Biomed Mater,2012,7(5): 054105.
    [66]Salgado AJ, Coutinho OPReis RL. Bone tissue engineering:state of the art and future trends[J]. Macromol Biosci,2004,4(8):743-65.
    [67]Shen Z, Kang C, Chen JJ, et al. Surface modification of polyurethane towards promoting the ex vivo cytocompatibility and in vivo biocompatibility for hypopharyngeal tissue engineering[J]. J Biomater Appl,2012.
    [68]Thibault RA, Mikos AGKasper FK. Scaffold/Extracellular matrix hybrid constructs for bone-tissue engineering[J]. Adv Healthc Mater,2013,2(1): 13-24.
    [69]Tripathi A, Kathuria NKumar A. Elastic and macroporous agarose-gelatin cryogels with isotropic and anisotropic porosity for tissue engineering[J]. J Biomed Mater Res A,2009,90(3):680-94.
    [70]Vitale-Brovarone C, Ciapetti G, Leonardi E, et al. Resorbable glass-ceramic phosphate-based scaffolds for bone tissue engineering:synthesis, properties, and in vitro effects on human marrow stromal cells[J]. J Biomater Appl,2011, 26(4):465-89.
    [71]Xie J, Blough ERWang CH. Submicron bioactive glass tubes for bone tissue engineering[J]. Acta Biomater,2012,8(2):811-9.
    [72]You Z, Bi X, Fan X, et al. A functional polymer designed for bone tissue engineering[J]. Acta Biomater,2012,8(2):502-10.
    [73]Masaeli E, Morshed M, Nasr-Esfahani MH, et al. Fabrication, characterization and cellular compatibility of poly(hydroxy alkanoate) composite nanofibrous scaffolds for nerve tissue engineering[J]. PLoS One,2013,8(2):e57157.
    [74]Santerre JP, Woodhouse K, Laroche G, et al. Understanding the biodegradation of polyurethanes:from classical implants to tissue engineering materials[J]. Biomaterials,2005,26(35):7457-70.
    [75]Kim KFisher JP. Nanoparticle technology in bone tissue engineering[J]. J Drug Target,2007,15(4):241-52.
    [76]Lopez-Perez PM, da Silva RM, Sousa RA, et al. Plasma-induced polymerization as a tool for surface functionalization of polymer scaffolds for bone tissue engineering:an in vitro study[J]. Acta Biomater,2010,6(9): 3704-12.
    [77]Fu S, Guo G, Gong C, et al. Injectable biodegradable thermosensitive hydrogel composite for orthopedic tissue engineering.1. Preparation and characterization of nanohydroxyapatite/poly(ethylene glycol)-poly(epsilon-caprolactone)-poly(ethylene glycol) hydrogel nanocomposites[J]. J Phys Chem B,2009,113(52):16518-25.
    [78]Fu S, Guo G, Wang X, et al. n-Hydroxyapatite/PCL-Pluronic-PCL Nanocomposites for Tissue Engineering. Part 2:Thermal and Tensile Study [J]. J Biomater Sci Polym Ed,2010.
    [79]Fu S, Guo G, Wang X, et al. Preparation and characterization of n-hydroxyapatite/PCL-pluronic-PCL nanocomposites for tissue engineering[J]. J Nanosci Nanotechnol,2010,10(2):711-8.
    [80]Jeong HS, Venkatesan JKim SK. Hydroxyapatite-Fucoidan Nanocomposites for Bone Tissue Engineering[J]. Int J Biol Macromol,2013.
    [81]Ghasemi-Mobarakeh L, Prabhakaran MP, Morshed M, et al. Electrospun poly(epsilon-caprolactone)/gelatin nanofibrous scaffolds for nerve tissue engineering[J]. Biomaterials,2008,29(34):4532-9.
    [82]Katti KS, Katti DRDash R. Synthesis and characterization of a novel chitosan/montmorillonite/hydroxyapatite nanocomposite for bone tissue engineering[J]. Biomed Mater,2008,3(3):034122.
    [83]Kawaguchi M, Fukushima T, Hayakawa T, et al. Preparation of carbon nanotube-alginate nanocomposite gel for tissue engineering[J]. Dent Mater J, 2006,25(4):719-25.
    [84]Guo F, Wang T, Chen A, et al. Bovine calcined bone for the repair of radial defect in a rabbit model[J]. J Tongji Med Univ,2000,20(3):242-5.
    [85]Tong G, Lei J, Zhong C, et al. The curative effect and safety of collagen sponge with different pH and content of protein implanted into orthopedic bone defect[J]. Sheng Wu Yi Xue Gong Cheng Xue Za Zhi,2012,29(6): 1125-30.
    [86]Liu JB, Lian W, Chen JT, et al. Diaphyseal defect repair with nacre/polylactic acid composite artificial bone in rabbits[J]. Di Yi Jun Yi Da Xue Xue Bao, 2004,24(9):1029-32,1036.
    [87]Knothe UR, Dolejs S, Matthew Miller R, et al. Effects of mechanical loading patterns, bone graft, and proximity to periosteum on bone defect healing[J]. J Biomech,2010,43(14):2728-37.
    [88]Qi X, Liu JGHuang LF. Experimental study in rabbit on repair of radial bone defect by bioactive degradable polymer[J]. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi,2001,15(4):202-5.
    [89]Xu YT, Gu JFShang P. Experimental study of bone repair induced by cryopreserved allograft periosteum and fetal bone composition in bone defect[J]. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi,2001,15(3):183-7.
    [90]Zhu W, Xiao J, Wang D, et al. Experimental study of nano-HA artificial bone with different pore sizes for repairing the radial defect[J]. Int Orthop,2009, 33(2):567-71.
    [91]Zhang HF, Zhao CY, Fan HS, et al. Histological and biomechanical study of repairing rabbit radius segmental bone defect with porous titanium[J]. Beijing Da Xue Xue Bao,2011,43(5):724-9.
    [92]Liu T, Zhang X, Li Z, et al. Management of chronic radial head dislocation associated with segment bone defect in ulna after osteomyelitis[J]. J Trauma Acute Care Surg,2012,73(4):1014-7.
    [93]Zhang W, Li G, Deng R, et al. New bone formation in a true bone ceramic scaffold loaded with desferrioxamine in the treatment of segmental bone defect:a preliminary study[J]. J Orthop Sci,2012,17(3):289-98.
    [94]Wu DJ, Hao AH, Zhang C, et al. Promoting of angiogenesis and osteogenesis in radial critical bone defect regions of rabbits with nano-hydroxyapatite/collagen/PLA scaffolds plus endothelial progenitor cells[J]. Zhonghua Yi Xue Za Zhi,2012,92(23):1630-4.
    [95]Fujioka M, Hayashida K, Murakami C, et al. Reconstruction of total nasal defect including skin, bone, and lining, using a single free radial forearm osteocutaneous perforator flap[J]. Plast Reconstr Surg,2012,129(5):854e-7e.
    [96]Niemeyer P, Szalay K, Luginbuhl R, et al. Transplantation of human mesenchymal stem cells in a non-autogenous setting for bone regeneration in a rabbit critical-size defect model[J]. Acta Biomater,2010,6(3):900-8.
    [97]Kim SJ, Jang JDLee SK. Treatment of long tubular bone defect of rabbit using autologous cultured osteoblasts mixed with fibrin[J]. Cytotechnology,2007, 54(2):115-20.
    [98]Glatt V, Kwong FN, Park K, et al. Ability of recombinant human bone morphogenetic protein 2 to enhance bone healing in the presence of tobramycin:evaluation in a rat segmental defect model[J]. J Orthop Trauma, 2009,23(10):693-701.
    [99]Sandor GK, Tuovinen VJ, Wolff J, et al. Adipose Stem Cell Tissue-Engineered Construct Used to Treat Large Anterior Mandibular Defect:A Case Report and Review of the Clinical Application of Good Manufacturing Practice-Level Adipose Stem Cells for Bone Regeneration[J]. J Oral Maxillofac Surg,2013.
    [100]Por YC, Barcelo CR, Salyer KE, et al. Bone generation in the reconstruction of a critical size calvarial defect in an experimental model [J]. Ann Acad Med Singapore,2007,36(11):911-9.
    [101]Kokubo S, Fujimoto R, Yokota S, et al. Bone regeneration by recombinant human bone morphogenetic protein-2 and a novel biodegradable carrier in a rabbit ulnar defect model[J]. Biomaterials,2003,24(9):1643-51.
    [102]Yu D, Li Q, Mu X, et al. Bone regeneration of critical calvarial defect in goat model by PLGA/TCP/rhBMP-2 scaffolds prepared by low-temperature rapid-prototyping technology[J]. Int J Oral Maxillofac Surg,2008,37(10): 929-34.
    [103]Kang SW, Bae JH, Park SA, et al. Combination therapy with BMP-2 and BMSCs enhances bone healing efficacy of PCL scaffold fabricated using the 3D plotting system in a large segmental defect model[J]. Biotechnol Lett,2012, 34(7):1375-84.
    [104]Hou R, Chen F, Yang Y, et al. Comparative study between coral-mesenchymal stem cells-rhBMP-2 composite and auto-bone-graft in rabbit critical-sized cranial defect model[J]. J Biomed Mater Res A,2007,80(1):85-93.
    [105]Rahman CV, Ben-David D, Dhillon A, et al. Controlled release of BMP-2 from a sintered polymer scaffold enhances bone repair in a mouse calvarial defect model[J]. J Tissue Eng Regen Med,2012.
    [106]Wu G, Liu Y, Iizuka T, et al. The effect of a slow mode of BMP-2 delivery on the inflammatory response provoked by bone-defect-filling polymeric scaffolds[J]. Biomaterials,2010,31(29):7485-93.
    [107]Stancoven BW, Lee J, Dixon DR, et al. Effect of bone morphogenetic protein-2, demineralized bone matrix and systemic parathyroid hormone (1-34) on local bone formation in a rat calvaria critical-size defect model[J]. J Periodontal Res,2013,48(2):243-51.
    [108]Canter HI, Vargel I, Korkusuz P, et al. Effect of use of slow release of bone morphogenetic protein-2 and transforming growth factor-Beta-2 in a chitosan gel matrix on cranial bone graft survival in experimental cranial critical size defect model[J]. Ann Plast Surg,2010,64(3):342-50.
    [109]Schwartz NDHicks BM. Eight-centimeter segmental ulnar defect treated with recombinant human bone morphogenetic protein-2[J]. Am J Orthop (Belle Mead NJ),2008,37(11):569-71.
    [110]Decker JF, Lee J, Cortella CA, et al. Evaluation of implants coated with recombinant human bone morphogenetic protein-2 and vacuum-dried using the critical-size supraalveolar peri-implant defect model in dogs[J]. J Periodontol,2010,81(12):1839-49.
    [111]Angle SR, Sena K, Sumner DR, et al. Healing of rat femoral segmental defect with bone morphogenetic protein-2:a dose response study[J]. J Musculoskelet Neuronal Interact,2012,12(1):28-37.
    [112]Brown KV, Li B, Guda T, et al. Improving bone formation in a rat femur segmental defect by controlling bone morphogenetic protein-2 release[J]. Tissue Eng Part A,2011,17(13-14):1735-46.
    [113]Tsuzuki N, Otsuka K, Seo J, et al. In vivo osteoinductivity of gelatin beta-tri-calcium phosphate sponge and bone morphogenetic protein-2 on an equine third metacarpal bone defect[J]. Res Vet Sci,2012,93(2):1021-5.
    [114]Luca L, Rougemont AL, Walpoth BH, et al. Injectable rhBMP-2-loaded chitosan hydrogel composite:osteoinduction at ectopic site and in segmental long bone defect[J]. J Biomed Mater Res A,2011,96(1):66-74.
    [115]Roohani-Esfahani SI, Dunstan CR, Davies B, et al. Repairing a critical-sized bone defect with highly porous modified and unmodified baghdadite scaffolds[J]. Acta Biomater,2012,8(11):4162-72.
    [116]Cui X, Zhang BXZhao DW. [Restoration of segmental bone defect by calcium sulfate pellet:experiment with rabbit][J]. Zhonghua Yi Xue Za Zhi,2009, 89(11):777-81.
    [117]Azad V, Breitbart E, Al-Zube L, et al. rhBMP-2 enhances the bone healing response in a diabetic rat segmental defect model[J]. J Orthop Trauma,2009, 23(4):267-76.
    [118]Seeherman HJ, Li XJ, Bouxsein ML, et al. rhBMP-2 induces transient bone resorption followed by bone formation in a nonhuman primate core-defect model[J]. J Bone Joint Surg Am,2010,92(2):411-26.
    [1]Ilmer M, Karow M, Geissler C, et al. Human osteoblast-derived factors induce early osteogenic markers in human mesenchymal stem cells[J]. Tissue Eng Part A,2009,15(9):2397-409.
    [2]Zhu H, Jiang XX, Wu Y, et al. [Identification of mesenchymal stem cells derived from rheumatoid arthritis synovial fluid and their regulatory effect on osteoblast formation] [J]. Zhongguo Shi Yan Xue Ye Xue Za Zhi,2009,17(4): 977-80.
    [3]Jiang SJ, He XS, Ni YD, et al. [Proliferation of the mesenchymal stem cells in a delayed fat flap:an experimental study in rabbits][J]. Zhonghua Zheng Xing Wai Ke Za Zhi,2009,25(4):287-9.
    [4]Bo B, Wang CYGuo XM. [Repair of cranial defects with bone marrow derived mesenchymal stem cells and beta-TCP scaffold in rabbits][J]. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi,2003,17(4):335-8.
    [5]Nather A, David V, Teng JW, et al. Effect of autologous mesenchymal stem cells on biological healing of allografts in critical-sized tibial defects simulated in adult rabbits[J]. Ann Acad Med Singapore,2010,39(8): 599-606.
    [6]Ruiz-Iban MA, Diaz-Heredia J, Garcia-Gomez I, et al. The effect of the addition of adipose-derived mesenchymal stem cells to a meniscal repair in the avascular zone:an experimental study in rabbits[J]. Arthroscopy,2011, 27(12):1688-96.
    [7]Dashtdar H, Rothan HA, Tay T, et al. A preliminary study comparing the use of allogenic chondrogenic pre-differentiated and undifferentiated mesenchymal stem cells for the repair of full thickness articular cartilage defects in rabbits[J]. J Orthop Res,2011,29(9):1336-42.
    [8]Lin CY, Chang YH, Lin KJ, et al. The healing of critical-sized femoral segmental bone defects in rabbits using baculovirus-engineered mesenchymal stem cells[J]. Biomaterials,2010,31(12):3222-30.
    [9]Kim SS, Song CK, Shon SK, et al. Effects of human amniotic membrane grafts combined with marrow mesenchymal stem cells on healing of full-thickness skin defects in rabbits[J]. Cell Tissue Res,2009,336(1):59-66.
    [10]Feng G, Zhao X, Liu H, et al. Transplantation of mesenchymal stem cells and nucleus pulposus cells in a degenerative disc model in rabbits:a comparison of 2 cell types as potential candidates for disc regeneration[J]. J Neurosurg Spine,2011,14(3):322-9.
    [11]Ohya M, Yamada Y, Ozawa R, et al. Sinus floor elevation applied tissue-engineered bone. Comparative study between mesenchymal stem cells/platelet-rich plasma (PRP) and autogenous bone with PRP complexes in rabbits[J]. Clin Oral Implants Res,2005,16(5):622-9.
    [12]Song YS, Ku JH, Song ES, et al. Magnetic resonance evaluation of human mesenchymal stem cells in corpus cavernosa of rats and rabbits[J]. Asian J Androl,2007,9(3):361-7.
    [13]Fujita H, Kurokawa K, Ogino T, et al. Effect of risedronate on osteoblast differentiation, expression of receptor activator of NF-kappaB ligand and apoptosis in mesenchymal stem cells[J]. Basic Clin Pharmacol Toxicol,2011, 109(2):78-84.
    [14]Abdallah BM, Haack-Sorensen M, Fink T, et al. Inhibition of osteoblast differentiation but not adipocyte differentiation of mesenchymal stem cells by sera obtained from aged females[J]. Bone,2006,39(1):181-8.
    [15]Sowa H, Kaji H, Canaff L, et al. Inactivation of menin, the product of the multiple endocrine neoplasia type 1 gene, inhibits the commitment of multipotential mesenchymal stem cells into the osteoblast lineage[J]. J Biol Chem,2003,278(23):21058-69.
    [16]Kulterer B, Friedl G, Jandrositz A, et al. Gene expression profiling of human mesenchymal stem cells derived from bone marrow during expansion and osteoblast differentiation[J]. BMC Genomics,2007,8(70.
    [17]Udehiya RK, Amarpal, Aithal HP, et al. Comparison of autogenic and allogenic bone marrow derived mesenchymal stem cells for repair of segmental bone defects in rabbits[J]. Res Vet Sci,2013.
    [18]Munisso MC, Kang JH, Tsurufuji M, et al. Cilomilast enhances osteoblast differentiation of mesenchymal stem cells and bone formation induced by bone morphogenetic protein 2[J]. Biochimie,2012,94(11):2360-5.
    [19]Wu X, Estwick SA, Chen S, et al. Neurofibromin plays a critical role in modulating osteoblast differentiation of mesenchymal stem/progenitor cells[J]. Hum Mol Genet,2006,15(19):2837-45.
    [20]Sun J, Zhou H, Deng Y, et al. Conditioned medium from bone marrow mesenchymal stem cells transiently retards osteoblast differentiation by downregulating runx2[J]. Cells Tissues Organs,2012,196(6):510-22.
    [21]Zhao JW, Gao ZL, Mei H, et al. Differentiation of human mesenchymal stem cells:the potential mechanism for estrogen-induced preferential osteoblast versus adipocyte differentiation[J]. Am J Med Sci,2011,341(6):460-8.
    [22]Sethi S, Radio NM, Kotlarczyk MP, et al. Determination of the minimal melatonin exposure required to induce osteoblast differentiation from human mesenchymal stem cells and these effects on downstream signaling pathways[J]. J Pineal Res,2010,49(3):222-38.
    [23]Plaisant M, Fontaine C, Cousin W, et al. Activation of hedgehog signaling inhibits osteoblast differentiation of human mesenchymal stem cells[J]. Stem Cells,2009,27(3):703-13.
    [24]Dahl M, Syberg S, Jorgensen NR, et al. Adipose derived mesenchymal stem cells-Their osteogenicity and osteoblast in vitro mineralization on titanium granule carriers [J]. J Craniomaxillofac Surg,2013.
    [25]Zhu F, Guo GH, Huang S, et al. [The effect of bone marrow mesenchymal stem cells engraftment on lung tissue at early stage of smoke inhalation injury in rabbits][J]. Zhongguo Wei Zhong Bing Ji Jiu Yi Xue,2011,23(1): 18-20.
    [26]Zhao M, Zhou J, Li X, et al. Repair of bone defect with vascularized tissue engineered bone graft seeded with mesenchymal stem cells in rabbits[J]. Microsurgery,2011,31(2):130-7.
    [27]Nicolaidou V, Wong MM, Redpath AN, et al. Monocytes induce STAT3 activation in human mesenchymal stem cells to promote osteoblast formation[J]. PLoS One,2012,7(7):e39871.
    [28]Backesjo CM, Li Y, Lindgren U, et al. Activation of Sirtl decreases adipocyte formation during osteoblast differentiation of mesenchymal stem cells[J]. Cells Tissues Organs,2009,189(1-4):93-7.
    [29]Mathews S, Mathew SA, Gupta PK, et al. Glycosaminoglycans enhance osteoblast differentiation of bone marrow derived human mesenchymal stem cells[J]. J Tissue Eng Regen Med,2012.
    [30]Lim TY, Wang W, Shi Z, et al. Human bone marrow-derived mesenchymal stem cells and osteoblast differentiation on titanium with surface-grafted chitosan and immobilized bone morphogenetic protein-2[J]. J Mater Sci Mater Med,2009,20(1):1-10.
    [31]Laurencin CT, Khan Y, Kofron M, et al. The ABJS Nicolas Andry Award: Tissue engineering of bone and ligament:a 15-year perspective[J]. Clin Orthop Relat Res,2006,447(221-36.
    [32]Meyer U, Joos UWiesmann HP. Biological and biophysical principles in extracorporal bone tissue engineering. Part Ⅰ[J]. Int J Oral Maxillofac Surg, 2004,33(4):325-32.
    [33]Wiesmann HP, Joos UMeyer U. Biological and biophysical principles in extracorporal bone tissue engineering. Part Ⅱ[J]. Int J Oral Maxillofac Surg, 2004,33(6):523-30.
    [34]Liebschner MA. Biomechanical considerations of animal models used in tissue engineering of bone[J]. Biomaterials,2004,25(9):1697-714.
    [35]Jose MV, Thomas V, Xu Y, et al. Aligned bioactive multi-component nanofibrous nanocomposite scaffolds for bone tissue engineering[J]. Macromol Biosci,2010,10(4):433-44.
    [36]Jose MV, Thomas V, Johnson KT, et al. Aligned PLGA/HA nanofibrous nanocomposite scaffolds for bone tissue engineering[J]. Acta Biomater,2009, 5(1):305-15.
    [37]Delabarde C, Plummer CJ, Bourban PE, et al. Biodegradable polylactide/hydroxyapatite nanocomposite foam scaffolds for bone tissue engineering applications[J]. J Mater Sci Mater Med,2012,23(6):1371-85.
    [38]Bhattacharyya S, Kumbar SG, Khan YM, et al. Biodegradable polyphosphazene-nanohydroxyapatite composite nanofibers:scaffolds for bone tissue engineering[J]. J Biomed Nanotechnol,2009,5(1):69-75.
    [39]Duan B, Wang M, Zhou WY, et al. Three-dimensional nanocomposite scaffolds fabricated via selective laser sintering for bone tissue engineering[J]. Acta Biomater,2010,6(12):4495-505.
    [40]Kruyt MC, van Gaalen SM, Oner FC, et al. Bone tissue engineering and spinal fusion:the potential of hybrid constructs by combining osteoprogenitor cells and scaffolds[J]. Biomaterials,2004,25(9):1463-73.
    [41]Cui H, Liu Y, Deng M, et al. Synthesis of biodegradable and electroactive tetraaniline grafted poly(ester amide) copolymers for bone tissue engineering[J]. Biomacromolecules,2012,13(9):2881-9.
    [42]de Oliveira AA, de Carvalho SM, Leite Mde F, et al. Development of biodegradable polyurethane and bioactive glass nanoparticles scaffolds for bone tissue engineering applications[J]. J Biomed Mater Res B Appl Biomater,2012,100(5):1387-96.
    [43]Duan BWang M. Customized Ca-P/PHBV nanocomposite scaffolds for bone tissue engineering:design, fabrication, surface modification and sustained release of growth factor[J]. J R Soc Interface,2010,7 Suppl 5(S615-29.
    [44]El-Ghannam A, Ning CQMehta J. Cyclosilicate nanocomposite:a novel resorbable bioactive tissue engineering scaffold for BMP and bone-marrow cell delivery[J]. J Biomed Mater Res A,2004,71(3):377-90.
    [45]Fernandez JM, Molinuevo MS, Cortizo MS, et al. Development of an osteoconductive PCL-PDIPF-hydroxyapatite composite scaffold for bone tissue engineering[J]. J Tissue Eng Regen Med,2011,5(6):e126-35.
    [46]Fricain JC, Schlaubitz S, Le Visage C, et al. A nano-hydroxyapatite--pullulan/dextran polysaccharide composite macroporous material for bone tissue engineering[J]. Biomaterials,2013, 34(12):2947-59.
    [47]Hankemeier S, van Griensven M, Ezechieli M, et al. Tissue engineering of tendons and ligaments by human bone marrow stromal cells in a liquid fibrin matrix in immunodeficient rats:results of a histologic study[J]. Arch Orthop Trauma Surg,2007,127(9):815-21.
    [48]Kheir E, Stapleton T, Shaw D, et al. Development and characterization of an acellular porcine cartilage bone matrix for use in tissue engineering[J]. J Biomed Mater Res A,2011,99(2):283-94.
    [49]Kim HJ, Kim UJ, Vunjak-Novakovic G, et al. Influence of macroporous protein scaffolds on bone tissue engineering from bone marrow stem cells[J]. Biomaterials,2005,26(21):4442-52.
    [50]Lacroix D, Chateau A, Ginebra MP, et al. Micro-finite element models of bone tissue-engineering scaffolds[J]. Biomaterials,2006,27(30):5326-34.
    [51]Wu DJ, Hao AH, Zhang C, et al. [Promoting of angiogenesis and osteogenesis in radial critical bone defect regions of rabbits with nano-hydroxyapatite/collagen/PLA scaffolds plus endothelial progenitor cells][J]. Zhonghua Yi Xue Za Zhi,2012,92(23):1630-4.
    [52]Niemeyer P, Szalay K, Luginbuhl R, et al. Transplantation of human mesenchymal stem cells in a non-autogenous setting for bone regeneration in a rabbit critical-size defect model[J]. Acta Biomater,2010,6(3):900-8.
    [53]Kim SJ, Jang JDLee SK. Treatment of long tubular bone defect of rabbit using autologous cultured osteoblasts mixed with fibrin[J]. Cytotechnology, 2007,54(2):115-20.
    [54]Niemeyer P, Szalay K, Luginbuhl R, et al. Transplantation of human mesenchymal stem cells in a non-autogenous setting for bone regeneration in a rabbit critical-size defect model[J]. Acta Biomater,2010,6(3):900-8.
    [55]Kim SJ, Jang JDLee SK. Treatment of long tubular bone defect of rabbit using autologous cultured osteoblasts mixed with fibrin[J]. Cytotechnology, 2007,54(2):115-20.
    [56]Glatt V, Kwong FN, Park K, et al. Ability of recombinant human bone morphogenetic protein 2 to enhance bone healing in the presence of tobramycin:evaluation in a rat segmental defect model[J]. J Orthop Trauma, 2009,23(10):693-701.
    [57]Sandor GK, Tuovinen VJ, Wolff J, et al. Adipose Stem Cell Tissue-Engineered Construct Used to Treat Large Anterior Mandibular Defect:A Case Report and Review of the Clinical Application of Good Manufacturing Practice-Level Adipose Stem Cells for Bone Regeneration[J]. J Oral Maxillofac Surg,2013.
    [58]Por YC, Barcelo CR, Salyer KE, et al. Bone generation in the reconstruction of a critical size calvarial defect in an experimental model[J]. Ann Acad Med Singapore,2007,36(11):911-9.
    [59]Kokubo S, Fujimoto R, Yokota S, et al. Bone regeneration by recombinant human bone morphogenetic protein-2 and a novel biodegradable carrier in a rabbit ulnar defect model[J]. Biomaterials,2003,24(9):1643-51.
    [60]Yu D, Li Q, Mu X, et al. Bone regeneration of critical calvarial defect in goat model by PLGA/TCP/rhBMP-2 scaffolds prepared by low-temperature rapid-prototyping technology[J]. Int J Oral Maxillofac Surg,2008,37(10): 929-34.
    [61]Kang SW, Bae JH, Park SA, et al. Combination therapy with BMP-2 and BMSCs enhances bone healing efficacy of PCL scaffold fabricated using the 3D plotting system in a large segmental defect model[J]. Biotechnol Lett, 2012,34(7):1375-84.
    [62]Hou R, Chen F, Yang Y, et al. Comparative study between coral-mesenchymal stem cells-rhBMP-2 composite and auto-bone-graft in rabbit critical-sized cranial defect model[J]. J Biomed Mater Res A,2007, 80(1):85-93.
    [63]Rahman CV, Ben-David D, Dhillon A, et al. Controlled release of BMP-2 from a sintered polymer scaffold enhances bone repair in a mouse calvarial defect model[J]. J Tissue Eng Regen Med,2012.
    [64]Wu G, Liu Y, Iizuka T, et al. The effect of a slow mode of BMP-2 delivery on the inflammatory response provoked by bone-defect-filling polymeric scaffolds[J]. Biomaterials,2010,31(29):7485-93.
    [65]Stancoven BW, Lee J, Dixon DR, et al. Effect of bone morphogenetic protein-2, demineralized bone matrix and systemic parathyroid hormone (1-34) on local bone formation in a rat calvaria critical-size defect model[J]. J Periodontal Res,2013,48(2):243-51.
    [66]Canter HI, Vargel I, Korkusuz P, et al. Effect of use of slow release of bone morphogenetic protein-2 and transforming growth factor-Beta-2 in a chitosan gel matrix on cranial bone graft survival in experimental cranial critical size defect model[J]. Ann Plast Surg,2010,64(3):342-50.
    [67]Schwartz NDHicks BM. Eight-centimeter segmental ulnar defect treated with recombinant human bone morphogenetic protein-2 [J]. Am J Orthop (Belle Mead NJ),2008,37(11):569-71.
    [68]Decker JF, Lee J, Cortella CA, et al. Evaluation of implants coated with recombinant human bone morphogenetic protein-2 and vacuum-dried using the critical-size supraalveolar peri-implant defect model in dogs[J]. J Periodontol,2010,81(12):1839-49.

© 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