新型可吸收材料PLLA/PLLA-gHA对骨折愈合相关蛋白表达影响的实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
金属材料是骨科、颅颌面外科骨折常用的内固定材料,但在体内长期应用会存在一系列并发症。人们一直在寻找更完善的固定系统,目前聚乳酸(PLA)高分子生物可吸收第二代内固定材料成为研究热点。其中在聚乳酸材料中引进羟基磷灰石(HA)材料制成的PLA/ HA复合材料得到普遍肯定,但在植入人体后,其初始力学强度的在短时间内迅速降低,同时对周围组织产生不同程度的炎性反应,限制其在临床应用。
     我们的合作单位中国科学院长春应用化学研究所研制出的具有自主知识产权的新型可吸收PLLA/PLLA-gHA骨修复材料,本课题组前期实验表明该复合材料具有良好的生物相容性,与传统生物可降解材料相比更能提高初始力学强度,而且可以提供较比稳定的内环境,对骨折愈合具有促进作用。骨折愈合过程由全身因子与局部因子共同调节,包括骨形态发生蛋白(BMPs)、胰岛素样生长因子-I(IGF-I)、血管内皮细胞生长因子(VEGF)、转化生长因子(TGF-β1)、成纤维细胞生长因子(FGF)、血小板衍生生长因子(PDGF)在内的生长因子参与了骨细胞的增殖、分化以及间充质的合成,同时作为信号物质参与骨折修复。I型胶原基因的表达是钙结节形成和成骨细胞表型成熟的基本保障,可以促进成骨细胞分化、增强成骨细胞粘附能力。在骨折早期起重要促进骨折愈合作用。新型复合材料PLLA/PLLA-gHA是否通过影响骨折断端生长因子的表达而促进缩短骨折愈合时间,目前研究尚不明确。本研究利用PLLA/PLLA-gHA复合材料固定于比格犬下颌骨骨折模型,通过免疫组化以及western-blot方法检测TGF-β1、BMP2、bFGF、VEGF、IGF-I、PDGF生长因子和I型胶原的表达和分布,探讨促进骨折愈合的机制。结果表明PLLA/PLLA-gHA复合材料在骨折愈合过程中可以促进TGF-β1、BMP2、bFGF、VEGF、IGF-I、PDGF和I型胶原表达,加快骨钙化,从而加速骨折愈合,为临床应用提供有效的理论基础。
Bone fracture is a common injury in the clinical practice of the orthopedics and cranio-maxillo-facial surgery. The healing of fractured bone is a complex physiological process, which includes vascular migration, angiogenesis and callus remodeling. Additionally, it involves the mediation of the differentiation and proliferation of cells by cell growth factor, as well as the effects of cell growth factor in the bone induction and molecular signal transduction. It is a multi-serial repairing process. In the management of patients with bone fracture, the traditional method is to use metal alloy as the internal fixer. The advantage of such material includes high mechanical strength and torsional modulus. However, it has the following limitations: The implantation of the metal fixer into the body may lead to electrolytic corrosion and subsequent release of metal ion. The later can cause inflammatory response of the local tissue, which thereby prompts a second surgery; the mismatch of bone rigidity and metal rigidity, which may lead to the absorption and weakening of the cortex bone underneath the internal fixation, thereby retarding the healing process. The high rigidity may increase the risk of another fracture of the reconstructed bone; T he metal internal fixer may induce the formation of fibrous cyst in the bone, which can separate the fixer and bone; The metal fixer must be removed after fracture healing to prevent bone atrophy (underneath the splint) due to lack of functional stimulation and inflammatory pain caused by the loosening and corrosion of metal instruments. Due to the limitations, many investigators have engaged in the development of absorbable bone fixation material. The emergence of bioabsorbable material allows investigators to establish a more accomplished internal fixation system. As the absorbable polymers materials do not require a second operation to remove, they are especially suitable for patients who only needs temporary implantation.
     The most extensively investigated material is poly Lactic acid (PLA); PLA is thermoplastic polyester and is made from fomented products or lactic acid of the plant stem and root. It is free of toxicity and stimulation, and has good bio-compatibility. The Food and Drug Admistration of USA labels it is a safe product. The mechanical strength is reduced by degradation, and stress is slowly shifted to the site of fracture, which can stimulate the healing process. Moreover, the metal covering effect during the use of metal fixer can be avoided, thereby, ameliorating psychological and physiological stresses of patients. The material can be completely degraded, absorbed and excreted by body. Thus, the second operation to remove the fixer is avoided. However, there are several limitations in its uses: i) the mechanical strength, rigidity and elastic modulus are all lower than natural bone cortex, thus, it can only be used for the non-weight bearing fracture sites. And the strength of such material needs to be improved. ii) Sterile inflammation may complicate the healing process during the material degradation.
     In an attempt to solve this problem, Chang Chun institute of applied chemistry Chinese academy of sciences developed the new PLLA/PLLA-gHA material (with independent intellectual property) for bone repairing. This technique has firstly performed grafting modification to gHA with the PLLA, the resultant complex PLLA-gHA is then mixed with PLLA to form the PLLA/PLLA-gHA mixture. HA is also called biological ceramics, which is similar to the bone matrix in the structure and composite, and has optimal tissue compatibility and effects of bone induction and conduction. We have conducted studies to assess the bio-compatibility of the mixture, the results showed that the PLLA/PLLA-gHA composite is free of cytotoxicity and acute toxicity that would not cause hemolytic reaction and rejection. It has biological safety and meets the relevant criteria. Compared with PLA material, it has higher mechanical strength and delayed degradation process, which satisfies the bone fracture fixation requirement of the beagle mandible. This material can improve the toughness to meet the mechanical requirement of fracture fixation. It can promote the adhesion and proliferation of the mouse embryo osteoblasts, and promote the osteoblasts to secrete extracellular matrix and form calcium nodules, which shows optimal compatibility with osteoblasts.
     With the development of molecular biology, various cell growth factors that have impact on the bone fracture healing have been discovered in recent years. And the understanding of fracture healing has transformed from histological level to the cellular and molecular levels. The initial phase of the study showed that, as compared with pure PLA material, PLLA/PLLA-gHA has optimal bone conduction property. However, the expressions of growth factors during the degradation of this material remain to be investigated. The growth factors are proteins of small molecular weight. Despite the small concentrations in the tissues and blood, the growth factors play important roles in the cell mitosis, matrix synthesis and tissue differentiation. The present study detected the expressions and distributions of TGF-β1, BMP2, bFGF, VEGF, IGF-I and PDGF by using immunohistochemical assay. And the expressions and distributions of typeⅠcollagen in callus were detected using Western blot method. The result will provide theoretical implications for clinical practice.
     The experiment was divided into three stages:
     (1) Beagle canine model of mandible fracture was established, which was fixed using PLLA/PLLA-gHA plate (experimental group) and titanium plate (control group). The canines were sacrificed at 1, 2, 3,4, 6,8 and 12 weeks after operation. The callus was obtained and sectioned for pathological examination. The sections were fixed, de-calcified, embedded and HE stained.
     (2) The modeling and sample harvesting were similar to the above-mentioned. The expressions and distributions of TGF-β1, BMP2, bFGF, VEGF, IGF-I and PDGF were detected by using immunohistochemical assay. The inter-group comparisons were performed to assess the mechanical property of PLLA/PLLA-gHA composite and its impact on the bone fracture healing.
     (3) The modeling and sample harvesting were similar to the above-mentioned. The expressions and distributions of CollageⅠin callus were detected using Western blot method.
     The results and conclusions of the study are as follows:
     (1) Compared with the experimental group and the control group, there was more fibroblasts increased in the experimental group, a large number of cartilage cells and osteoblasts proliferated, trabecular bone and angiogenesis appeared after 1 W; after 2W, endochondral bone formation in experimental group was more than in the control group, a large number of osteoblast proliferated, bone callus rich in blood vessels; after 3W: the experimental group was rich in osteoclasts and osteoblasts, new trabecular bone formed and cartilage formation was more than in the control group, bone salts deposited in cartilage matrix, angiogenesis was obvious, part of bone callus crossed the fracture gap, rich in blood vessels; after 4W, the formation of osseous callus and fibrous callus in the experimental group were more than the control group, the fracture gap was filled with new bone, more Haversian canal can be found; after 6W, osteoblasts and bone canaliculus in the experimental group was more than in the control group; after 8W, in the experimental group Haversian canal reduced in the fracture gap, new bone maturation appeared, bone cells arranged in concentric circles , bone lacunae arranged in neat rows, external callus is very small in amount; control group showed extensive bone formation, osteoblasts was in a random arrangement, fracture gap was connected by newly formed bone, The number of Lacuna is less than the experimental group; after 12W: fracture line disappeared and the difference between the experimental group and the control group decreased.
     (2) TGF-β1 protein was mainly expressed in osteoblasts, chondrocytes, and mesenchymal cells, the positive indexes in the postoperative experimental group after 2W, 3W, 4W were greater than the control group, and there was a significant difference (p <0.01 ), peak positive indexes in two groups appeared after 2W, then decreased gradually to no expression after 8w. Other time points no significant difference (p> 0.05).
     (3) BMP-2 protein was expressed in osteoblasts, chondrocytes, mesenchymal cells and bone matrix, peak positive indexes in two groups appeared after 2W, then decreased gradually to no expression after 12w. There was a very significant difference after 2W when compared the tow groups(p <0.01), after 3W, 4W there was a significant difference (p <0.05), the other time points no significant difference (p> 0.05).
     (4) bFGF protein was mainly expressed in osteoblasts, cartilage cells and bone matrix, the positive index began to rise since the 2w, 4w to peak, then decreased gradually to 12w when the negative expression showed. There was a very significant difference after 4W when compared the tow groups(p <0.01). Aafter 2W,3W there was a significant difference (p <0.05), the other time points no significant difference (p> 0.05).
     (5) VEGF protein mainly expressed in endothelial cells, cartilage cells, bone cells and osteoblasts, peak positive indexes in the two groups appeared after the 3W, then decreased gradually to no expression after 8w. Positive cell indexes in the experimental group after 2W, 4W, showed a significant difference than the control group (p <0.05), there was a very significant difference after 3W (p <0.01), the other time points difference was not statistically significant (p> 0.05).
     (6) PDGF protein was mainly expressed in mesenchymal cells, bone matrix, endothelial cells, cartilage cells and osteoblasts. In the experimental group after 1W, 2W,3W compared the two groups there was a significant difference (p <0.05),The peak value is located at 1W, the other time points no significant difference (p > 0.05).
     (7) IGF-1 protein mainly expressed in endothelial cells, osteoblasts and chondrocytes, peak positive index appeared at 2W, then decreased gradually to 12w when no expression appeared. Positive cell indexes in the experimental group after operation 1W, 2W, 3W were more than in the control group, which after 2W, 3W, compared the two groups there was a significant difference (p <0.01), after 1W there was a significant difference (p <0.05), the other time points no significant difference (p> 0.05).
     (8) Effect on type I collagen: the expression of type I collagen was weak in both groups after 1W, after 2W, expression levels in both groups increased, and the experimental group and the control group appeared significant difference (p <0.05). After 3W expression levels reached the peak in both groups and there was a significant difference between two groups (p <0.01). The expression of type I collagen in the two groups after4W was a significantly decreased, but the experimental group compared with the control group, there was still a significant difference (p <0.05). After 6W and 8W a slight expression of type I collagen appeared in both group, difference was not statistically significant (p> 0.05). There were no postoperative expression of type I collagen after 12W.
     Conclusions: the new bone repairing material PLLA/PLLA-gHA can satisfy the requirement of fracture fixation of the beagle mandible and can promote the healing of the mandible fracture of Beagles. Moreover, the protein expressions of TGF-β1, BMP2, bFGF, VEGF, IGF-I, PDGF and CollageⅠcan be significantly increased. The results provide biological basis for other basic researches of clinical application, thereby accelerating the early clinical use of the new material.
引文
[1] Rokkanen PU, Bostman O, Patiala H, et al. Bioabsorbabl fixation in orthopaedic surgery and traumatology. Biomaterials. 2000:21(24):2607-2613.
    [2]任杰,周新宇.羟基磷灰石/聚乳酸及其共聚物复合生物材料[J].上海生物医学工程, 2004,25(1):46-50.
    [3]金丹,斐国献,王前.骨髓基质细胞与生物活性玻璃陶瓷和聚乳酸生物相容性的实验研究[J].中国修复重建外科杂志, 2000,1(14):39-43.
    [4] P. Tormala, et al. J. Biomed Mater Res. 1991,25:1.
    [5] Bosman OM. Current concepts review absorbable implants for fixation of fracture [J]. J Bone Joint Surg,1991,73(2):148.
    [6]史铁钧,董智贤.聚乳酸的性能、合成方法及应用[J].化工新型材料, 2001,29(5):13-16.
    [7] Hollinger JO, Brekke J, Cruskin E, et al. Role of bone sustitutes[J]. Clin Orthop, 1996,(324):55-65.
    [8] Hideto Tsuji, Shinya Miyauehi. Enzymatiz Hydrolysis of free and Restrieted Amorphoas Regions in POly(L-laetide) Films with Different Crystallinities and a Fired Crystalline Thickness. Polymer, 2001,42:4463.
    [9]陈宇轩,吕春堂,雷德林,等.生物可吸收材料L/DL聚乳酸的生物相容性及体内降解[J].第四军医大学学报, 2000,21(4):417-420.
    [10] Galgut P, Pitrola R, Waite l, et al. Histological evaluation of biodegradable and non-biodegradable membranes placed transcutaneously in rats[J]. Clin Periodontol, 1991,18,581-586.
    [11] Vainionpaa SI, Vihtonen K. Bioabsorbable fixation in orthopaedic surgery and traumatology. Biomaterials, 2000,21(24):2607-2613.
    [12] Bos R, Rozema A, Boering G,et al. Degradation and tissue reaction to biodegradable poly(L-lactide) for use as internal fixation of fracture. Biomaterials, 1991,12:32.
    [13] Friend G, Grace K, Stone HA. L-osteotomy with absorbable fixation for correctionof tailor’s bunion[J]. J Foot Ankle Surg, 1993,32(1):14-9.
    [14] Weiler A, Helling HJ, Kirch U, et al. Foreign-body reaction and the coueseof osteolydid after polyglycolide implante for fracture fixation. Experimental study in sheep[J]. J Bone Joint Surg, 1996,78B:469-476.
    [15] Y. Matsusue, et al. Clin. Orthop, 1995,317:246.
    [16] M. V. D. Elst, et al. J. Biomed. Mater. Res, 1996,30:139.
    [17] Ashammakhi N. Neomembranes: a concept review with special reference to self-reinforced polyglycolide membranes.J Biomed Mater Res(Appl Biomater) 1996,33:297-303.
    [18] Hollinger JO. Biodegradable bone repair materials. Synthetic polymers and ceramics. Clin Orthop, 1986,207:290-305.
    [19] Hammerle CH, Schmid J, Lang NP, et al. Temporal dynamics of healing in rabbit cranialdefects using guided bone regeneration. J Oral Maxillofac Surg 1995,53:167-174.
    [20] Honllinger JO, Brekke J, Cruskin E, et al. Expermental study on glycol acid [J]. Gan To Kagaku aryoho. 1995:22(11)1641-3.
    [21]胡韶楠,顾玉东.可吸收聚合材料髓内针在手外科的应用.中华手外科杂志, 1995,11(3):179.
    [22] Bozic KJ, Perez LE, WiLson DR, et al. Mechanical testing of bioresorbable implants for use in metacarpal fracture fixation. J Hand Surg. 2001,26:755-761.
    [23] Waris E, Ashammakhi N, Happonen H, et al. Bioabsorbable miniplating versus metallic fixation for metacarpal fractures. Clin Orthop Relat Res. 2003,410:310-319.
    [24] Rokkanen P, Bostman O, Vainionpaa S, et al. Absorbable devices in the fixation of fractures. J Trauma, 1996,40(3 suppl):123.
    [25] Weiler A, Helling HJ, Kirch U, et al. Foreign-body reaction and the course of osteolysis after polyglycolide implants for fracture fixation.J Bone Joint Surg Br, 1996,78(3):369.
    [26] Albert RA Dijkema, Maarten van der Elst, Roelf S Breed-erveld, et al. Surgical treatment of fracture-dislocations of the ankle joint with biodegradable implants: a prospective randomized study. J Trauma, 1993,34:82.
    [27] Suuroen R, Mannien MJ, Pohjonen T, et al. Mandibular osteotomy fixed with biodegradable plate and screws:an animal study BrJ Oral Maxillofac Surg, 1997,35(5):341-348.
    [28] Kallella I, Laine P, Sunonen R, et al. Skeletal stability follow in mandibular advancement and rigid fixation with polylactide biodegradable screw[J]. Int J Oral Maxillofac Surg 1998,27:3-8.
    [29] Kallela I, Iizuka T, Salo A, Lindqvist C: Lag screw fixation of anterior mandibular fractures using biodegradable polylactide screws: a preliminary report. J Oral Maxillofac Surg 1999,57:113-118. A clinical report of 11 patients in whom a new application(lag screw fixation)of resorbable technology was used.The results are promising and point to the expanding future for these devices.
    [30] Vert M, Christel P, Chabot F, et al. Macromolecular biomaterial Boca Raton. CRC Press, 1984,119-142.
    [31] Werner A, Wild A, Ilg A, Krauspe R. Secondary intra-articular dislocation of a broken bioabsorbable interference screw after anterior cruciate ligament reconstruction[J]. Knee Surg Sports Traumatol Arthrosc. 2002 Jan, 10(1):30-2.
    [32]牛金龙,张镇西,蒋大宗.多孔羟基磷灰石生物陶瓷的合成和特性研究进展,生物医学工程学杂志. 2002,19:302-305.
    [33]李世普,编著,生物医用材料导论.武汉:武汉工业大学出版社, 2000.214-218.
    [34] Wei Jie, Li Yubao. Tissue engineering scaffold material of nano-apatite crystals and polyamide composite[J]. European Polymer Journal, 2004,40,509-515.
    [35] Du C, Cui FZ, Zhu XD, de Groot K. Three-dimensional nano-Hap/collagen matrix loading with osteogenic cells in organ culture.J Biomed Mater Res, 1999,44(4):407-412.
    [36] WebsteTJ, ErgunC, Doremus RH, et al. Biomaterials, 2000,21(13):1803-1810.
    [37] Jarcho M. Retrospective analysis of hydroxyapatite development for oral implant applications[J]. Dent Clin North Am, 1992,36(1):19-26.
    [38] Yoshikawa T, Ohgushi H, Tamai S.Immediate bone forming capability of prefabricated osteogenic hydroxyapatite [J]. J Biomed Mater Res, 1996,32(3):481-485.
    [39] Ripamonti U, Ceook J, Kirkbride AN. Sintered porous hydroxyapatites withintrinsic osteoinductive activity:geometric induction of bone formation.South African J Sci, 1999,95:335.
    [40] Yuan H, Kurashina K, de Bruijn JD, Li Y, de Groot K, Zhang X. A preliminary study on oseoinduction of two kind of calcium phosphate ceramic. Biomaterials, 1999,20:1799-1806.
    [41] Saadeh PB, Khosla RK, Mehrara BJ, Steinbrech DS, Mccormick SA, DeVore DP, Longaker MT. Repair of a critical size defect in the rat mandible using allogenic type I collagen. J Graniofac Surg, 2001,12:573-579.
    [42]兰建宏,鲁步超,寇强勇,等. HA-TI种植体修复牙列缺损疗效的观察[J].中国口腔种植学杂志, 1997,2:134.
    [43] Alliot-Lieht B, De Lange GL, Gregoire M. Effects of hydroxyapatite partieles onPeriodontal ligament fibroblast-liked cell behavior[J]. J Per iodontol, 1997,68(2):158-165.
    [44] Laurenein CT,Attawia MA,Elgendy HE,et al.Tissue engineered bone regeneration using degradable Polrners.The formulation of mineralized matriees[J]. Bone,1996(l):93-99.
    [45] Hong Z, Qiu X, Sun J, et al. Grafting polymerization of L-lactide on the surface of hydroxyapatite nano-crystals. [J]. PoLymer, 2004,45:6699-6706.
    [46] Hong ZK, Zhang PB, He CL, et al. Nano-composite of poly (L-Lactide) and surface grafted hydroxyapatite: Mechanical properties and biocompatibility. Biomaterials 2005,26(32),6296-6304.
    [47]邵英,路来金,荣莉等.聚L-乳酸/聚L-乳酸接枝的羟基磷灰石复合材料的生物相容性.吉林大学学报(医学版), 2007,33(1):57-60.
    [48] UematsuS, Mogi M, Deguchi T. Interleukin(IL)-l beta, IL-6, Tumor necr-osis Factor-a, epidermal growth factor,and beta2-microglobulin levels are el-evated in gingival crevicular fluid during human orthodontic tooth moveme-nt[J]. J Dent Res, 1996,75(1):562-567.
    [49] Filvaroff E, Erlebacher A, Ye J, et al. Inhibition of TGF-beta receptor si-gnaling in osteoblasts leads to decreased bone remodeling and inereased tra-beeular bone mass[J]. Develo ment, 1999,126(19):4267-4279.
    [50] ShimizuT, Mehdi R, YoshimuraY, et al. Sequential expression of boneMorphogenetic Protein, tumor necrosis factor, and their receptor sin bone- Forming reaction after mouse femoral marrow ablation[J]. Bone, 1998,23(2):127-133.
    [51] GosPodarowiez D. Fibroblast growth factor.Chemieal structure and biologic function[J]. Clin Orthp Relat Res, 1990,257:231-48.
    [52] Mc Carthy TL, Ji C, Casinghino S, et al. Altemate signaling pathways selectively regulate binding of insulin-like growth factor I and IIon fetal rat bone cells[J], J Cell Biochem, 1998,68(4):446-456.
    [53] Fujii H, Kitazawa R, Maeda S, et al. Expression of Platelet-derived growth factor Proteins and their receptor alpha and beta mRNAs during fracture healing in the normal mouse[J]. Histoehem Cell Biol, 1999,112(2):131-138.
    [54] Cho T. J, Gerstenfeld L. C, Einhorn T. A, et al. Differential temporal expression of members of the transforming growth factor beta superfamily during murine fracture healing[J]. J Bone Miner Res, 2002,(5):13-20.
    [55] Rose F. R, Oreffo R. O. Bone tissue engineering: hope vs hype [J]. Biochem Biophys Res Commun, 2002,14(7):2921~2927.
    [56] Danis R. The aim of internal fixation[J]. Clin O nhop, 1979,138:23.
    [57] Ilizarov GA The tension-stress effection the genes and growth of tissues: part I[J]. Clin Orthop, 1989,238-249.
    [58] Triffitt JT Initiation and enhancement of bone formation: areview[J]. Acta Orthop scand, 1987,58:573.
    [59] Thomas A. E. The cell and molecular biology of fracture healing.Clin Orthop, 1998, (355):7.
    [60]裘法祖,主编,外科学(第七版).人民卫生出版社, 2009,727-728.
    [61] Joseph A, Buckwalter, MD, MS, et al.陈启明,梁国穗等译,骨科基础科学(第二版).人民卫生出版社, 2002,326-328.
    [62] Steinbrech DS, Mehrara BJ, Saaedh PB, et al. Hypoxia regulatesVEGF expression and cellular proliferation by osteoblast in vitro [J]. Plast Reconstr Surg, 1999,104:738-747.
    [63] Bouletreau PJ, Warren SM, Spector JA, et al. Hypoxia and VEGF up-regulate BMP-2 mRNA and protein expression in microvascular endothelial cells: implications forfracture healing. [J] Plastic & Reconstructive Surgery. 2002,109(7):2384-2397.
    [64] Jingushi S, Jogce ME, Bolander ME. The role of transforming growth factor betal and fibroblast growth factors in fracture repair [J]. J Bone Joint Surg(Br), 1993,75:285.
    [65] Bourque WT, Hall BK, Gross M. The presence of several growth factors during the stages of fractur repair. [J]. J Bone Joint Surg (Br), 1993,75:282.
    [66] Einhorn TA. The cell and molecular blology of fracture healing[J]. Clin orthop, 1998,(355S):S7-S21.
    [67] Koseki T, Gao Y, Okahashi N, et al. Role of TGF-beta family in osteoclastogenesis induced by RANKL [J]. Cellular Signalling. 2002,14(1):31-36.
    [68] Sabokbar A, Sun SG. Kudo O, Danks L, et al. Transforming growth factor-beta induces osteoclast formation in the absence of RANKL. [J] Bone, 2004,34(1):57-64.
    [69] Sells Galvin RJ, Gatlin CL,Horn JW, et al. TGF-beta enhances osteoclast differentiation in hematopoietic cell cultures stimulated with RANKL and M-CSF. [J] Biochem & Biophys Res Com. 1999,265(1):233-239.
    [70] Lovibond AC, Haque SJ, Chambers TJ, et al. TGF-beta-induced SOCS3 expression augments TNF-alpha-induced osteoclast forma-tion. [J] Biochemical & Biophysical Research Communications. 2003,309(4):762-767.
    [71] Fox SW, Haque SJ, Lovibond AC, et al. The possible role of TGF-beta-induced suppressors of cytokine signaling expression in osteo-clast/macrophage lineage commitment in vitro. [J] Journal of Im-munology. 2003,170(7):79-87.
    [72]郝永强,戴尅戎.骨折愈合过程中血管内皮细胞生长因子表达的实验研究.中华骨科杂志, 1999,19(11):683-686.
    [73] Gerstenfeld L. C, Cullinane D. M, Barnes G.L. et al. Fracture healing as a post-natal developmental process:molecular, spatial, and temporal aspects of its regulation[J]. J Cell Biochem, 2003,88(6):873-884.
    [74] William Axelrad T, Kakar S. New technologies for the enhancement of skeletal repair[J]. Injury, 2007,38:49-62.
    [75] Urist M. R. Bone: formation by autoinduction [J]. Science, 1965,150(3): 893~899.
    [76] WozneyJ. M, Rosen V, Celeste A. J, et al. Novel regulators of bone formation:molecular clones and activities[J]. Science 1988,242(5):1528-1534.
    [77] Varga AC, Wrana JL. The disparate role of BMP in stern cell biology[J]. Oncogene, 2005,24(37):5713.
    [78] Gambaro K, Aberdam E, Virolle T, et al. BMP 4 induces a Smad-dependent apoptotic cell death of mouse embryonic stem cell-derived neural pre-cursor differ[J] Cel1 Death Differ, 2006,13(7):1075.
    [79] Ahmed N, Sammons J, Khokher MA, et al. Effect of bone mor-phogenetic PROTEIN-5 on hematopoietic stem cells and cytokine production in normal human bone marrow stroma [J] Experimental Hematology. 2000,28(12):1495.
    [80]苏佳灿,王家林,张春才,成骨蛋白-1促进骨折愈合研究进展[J].中国矫形外科杂志, 2001,8(6):599-601.
    [81] PengY, Kang Q, Luo Q, et al. Inhibitor of DNA binding/differentiation helix-looP-helix proteins mediate bone morphogenetic Protein-induced osteoblast differentiation of mesenehymal stem cells[J]. J Biol Chem, 2004,279(31):32941.
    [82] TemaatMF, Blokhuis TJ, Bakker FC, et al. The role of bone morphoge-netic proteins in Bone healing[J]. Osteo Traurna Care, 2003,11:122-125.
    [83] Deckers MM, van Bezooijen RL, van der Horst G, et al. Bone morphogenetic proteins stimulate angiogenesis through osteoblast-derived vascular endothelial growth factor A. [J] Endocrinology. 2002,143(4):1545-53.
    [84] Linkhart TA, Mohan S, Baylink DJ. Growth factors for bone growth and repair: IGF TGF-beta and BMP. Bone, 1996,19:15.
    [85] Kubota K, Iseki S, Kurda S, et a1. Synergisitc effect of fibroblast growth factor-4 in ectopic bone formation induced by bone morpho-genetic protein-2[J]. Bone, 2002 31(4):465-471. 173-180.
    [86]谭龙益,孔宪涛.转化生长因子β受体的研究进展[J].国外医学临床生物化学与检验学分册, 1997,8(2):63-66.
    [87] Massague J. TGF-signal transduction [J]. AnnuRev Biochem, 1998,67(7): 753-791.
    [88] Piek E, Heldin CH, Dijke PT. Specificity, diversity, and regulation in TGF-βsuperfamily signaling[J]. FASEBJ, 1999,13(15):2105-2124.
    [89] Sandberg, M. M. Gene expression during bone repair[J]. Clin Orthop, 1993,289(7):292-312.
    [90] Assoian RK, et al. Typeβtransforming growth factor human platelets: release during platelet degranulation and action on vascu-lar smooth muscle cell. J Cell Biol, 1986,102.
    [91] Lieberman J. R., Daluiski A, T. A. Einhorn. The role of growth factors in the repair of bone. Biology and clinical applications[J]. J Bone Joint Surg, 2002,6(2):1032-1044.
    [92] Chen CG, Chen CL, Liu HN. Primary papular xanthoma of children: a clinicopathologic, immunohistopathologic and ultrastructural study[J]. J Biol Chem, 1997,19(6):596-601.
    [93]苏佳灿,张春才.骨折愈合中基因直接转入的作用机制[J].现代康复, 2001,5(1):62-63.
    [94] Ingram RT, Bonde SK, Riggs BL, et al. Effects of transforming growth factor beta and 1, 25-dihydroxy vitamin D3on the function, cytochemistry, and morphology of normal osteoblast-like cells[J]. Differentiation, 1994,55(2):153-163.
    [95] Liu P, Oyajobi BO, Russell RG, et al. Regulation of osteogenic differentiation of human bone marrow stromal cells:interaction between transforming growth factor-beta and 1, 25(OH)2vitamin D3in vitro[J]. Calcif Tissue Int, 1999,65(2):173-180.
    [96] Chen A, Davis BH, Sitrin MD, et al. Transforming growth factor-beta 1 signaling contributes to Caco-2 cell growth inhibition induced by 1, 25(OH)2D3[J]. Am J Physiol Gastrointest Liver Physiol, 2002,283(4):G864-874.
    [97] Monkawa T, Hiromura K, Wolf G, et al. The hypertrophic effect of transforming growth factor-beta is reduced in the absence of cyclin-de-pendent kinase-inhibitors p21 and p27[J]. J Am Soc Nephrol, 2002,13(5):1172-1178.
    [98] Ferrara N, Henzel WJ. Pituiary follicular cells secrete a novel heparin-binding growth factor specific for vascular endothelial cells. J Biochem Biophys Res Commun, 1989,161(2):851-855.
    [99] Senger DR, Gami SJ, Dvarak AM, et al. Tumor cells secrete a vascular permeability factor that promote accumulation of ascites fluid.Science, 1983,219:983-985.
    [100] Dvorak HF, Brown LF, Detmar M, et al. Vascular permeability factor/vascular endothelial growth factor, microvascular hyperpermeability and angiogenesis. Am J Pathol, 1995,146:1029-1039.
    [101] Ferrara N. Vascular endothelial growth factor: molecular and biological aspects. Curr Top Microbiol Immunol, 1999,237(1):1-3.
    [102] SNJ Sait, M Dougher-vermazen, TB show, et al. The kinase insert domain receptor gene(KDR)has relocated to chromosome 4q11-q12. Cytogenet cell Genet, 1995,70:145-146.
    [103] Waltenberger J, Claesso-welsh L, Siegbahn A, et al. Different signal transduction properties of KDR and FLT-1, two receptors for vascular endothelial growth factor. J Brol Chem, 1994,269:26988-26995.
    [104] Jeltsch M, Kaipainen A, Joukov V, et al. Hyperplasia of lymphatic vessels in VEGF-C transgenic mice.Science, 1997,276:1423-1425.
    [105] Soker S, Takashima S, Miao HQ, et al. Neuropilin-1 is expressed by endothelial and tumor cells as an isoform-specific receptor for vascular endothelial growth factor. Cell, 1998,92:735-745.
    [106]初同伟,王正国,朱佩芳,刘大维.骨折愈合过程中血管内皮生长因子及其受体的表达.中华创伤杂志. 2001,17(6)344-346.
    [107]陈希哲,杨连甲.血管内皮生长因子与骨的形成及修复.中华骨科杂志. 2001,21(7)441.
    [108] Hweiki D, Itin A, Soffer D, et al. VEGF induced by hypoxia may mediate hypoxia-initiated angiogenesis. Nature, 1992, 359,843-845.
    [109] Shifren TL, Doldi N, Ferrara N, et al. In the human fetus vascnlar endothelial growth factor is expressed in epithelial cells and mvocvtes, but not vascualar endothelium: Implkations for mode of action [J]. Clin Endocrinol Melab, 1994,79(7):316-322.
    [110] Mehrara BJ, Saaedh PB, Steinbrech DS, et al. Adenovirus-medi-ated gene therapy of osteoblasts in vitro and vivo[J]. J Bone Miner Res, 1999,14(8):1290-1301.
    [111] Saadeh PB, Mehrara BJ, et al. Transforming growth factor-betal modulated the expression of vascular endothelial growth factor by oesteoblasts. J Physiol, 1999,277(4):628-637.
    [112] Andrew JG, Hovland JA, Freemont AJ, et al. Platelet-Derived growth factor expression in normally healing human fractures [J]. Bone, 1994,15:203
    [113]苏佳灿,许硕贵,张春才.骨重建中细胞因子的作用及机制[J].中国矫形外科杂志, 2001,9(6):593-595.
    [114] Reilly JT. Pathogenesis and management of idiopathic myelofibrosis. Baillieres clin Haem atol 1998,11:751-767.
    [115] Mckinnon RD, Waldron S. PDGF alpha-receptor signal strength controls an RTK rheostat that integrates phosphoinositol 3 '-kinase and phospholipase Cgamma pathways during oligodendrocyte maturation [J]. JNeurosc,i 2005,6,25(14):3499-3508.
    [116] Sandilands E, Cans C, Fincham VJ, et al. RhoB and actin polymerization coordinate Src activation with endosome-mediated delivery to the membrane [J]. Dev Cell 2004,7(6):855-869.
    [117] Egan GG, Wainwright CL, Wadsworth RW, et al PDGF-induced signaling in proliferating and differentiated vascular smooth muscle: effects of altered intracellular Ca(2+) regulation[J]. Cardiovasc Res, 2005,6.
    [118] Oreffo R. Growth factors for skeletal reconstruction and fracture repair[J]. CurtOpin Investig Drugs. 2004,(4):419-423.
    [119] NashTJ, Howlett CR, Martin C, et al. Effect of platelet-derived growth factor on tibial osteotomies in rabbits. Bone, 1994,15:203
    [120] Platelet-derived growth factor and bone morphogenetic protein in the healing of mandibular fractures in rats[J]. Br J Oral Maxillofac Surg. 2003,41(3):173-178.
    [121] Fujii H, Kitazawa R, Maesas, et al. Sxpression of platelet-derived growth factor proteins and their receptor alpha and beta mRNA during fracture bealing in the normal mouser[J]. Histochem cell Bone, 1999,112(2):131-138
    [122] Masaya I, Yoshlaki A, Tomohiro O, et al. Effects of ultrasound and 1. 25-dihydroxyvitam in D3on growth factor secretion in co-cultures of osteob lasts and endothelial cells[J]. Ultrasound in Med & Bio, l 2000,26(1):161-166.
    [123] Gospodarowicz D. Purification of a fibroblast growth factor from bovinepituitary[J]. J Biol Chem, 1975,250:2515-2520.
    [124] Nakamura T, Hara Y, Tagawa M, et al. Recombinant human basic fibroblast growth factor accelerates fracture healing by enhancing callus remodelling in experimental dog tibial fractures[J]. J Bone Miner Res, 1998,13(9):942-949.
    [125] Suzuki A, Palmer G, Bonjour J P, et al. Stimulation of sodium-dependent phosphate transport and signaling mechanisms induced by basic fibroblast growth factor in MC3T3-E1 osteoblast-like cells[J]. J Bone Miner Res, 2000,15:95-102.
    [126] Tokuda H, Kozawa O, Uematsu T. Basic fibroblast growth factor stimulates vascular endothelial growth factor release in osteoblasts: divergent regulation by p42/p44 mitogen-activated protein kinase and p38 mitogen-activated protein kinase[J]. J Bone Miner Res, 2000,15:2371-2379.
    [127] Chaudhary L R, Hruska K A. The cell survival signal Akt is differentially activated by PDGF-BB, EGF, and FGF-2 in osteo-blastic cells[J]. J Cell Biochem, 2001,81:304-311.
    [128] Debiais F, Lefevre G, Lemonnier J. Fibroblast growth factor-2 induces osteoblast survival through a phosphatidylinositol 3-kinase -dependent, -beta-catenin-independent signaling pathway [J]. Exp Cell Res, 2004,297:235-246.
    [129] Tang C H, Yang R S, Huang T H, et al. Enhancement of fibronectin fibrillogenesis and bone formation by basic fibroblast growth factorviaprotein kinase C-dependent pathway in rat osteoblasts[J]. Mol Pharmacol, 2004,66:440-449.
    [130] Nakamura T, Hara Y, Tagawa M, et al. Recombinant human basic fibroblast growth factor accelerates fracture healing by enhancing callus remodeling in experimental dog tibial fracture J Bone Miner Res, 1998,13:942.
    [131] Nakamura T, Hanada K, Tamura M, et al. Stimulation of endosteal bone formation by systemic injections of reeombinant basic fibroblast growth factor in rat. Endocrinology, 1995,136:1276.
    [132] Hammerman MR. The growth hormone-insulin-like growth factor axis in kidney. Am J Physiol. 1989,257(4):F503.
    [133] Fan J, Wojnar MM, Theodorakis M, et al. Regulation of insulin-like growthfactor(IGF)-1 mRNA and peptide and IGF-binding proteins by interleukin-1. Am J Physiol, 996,270(3):R621.
    [134] Hill PA, Reyndds JJ, Meidle MC. Osteoblasts mediate insulin-like growth factor-Ⅰand-ⅡStimulation of Osteoblast foribation and function [J]. Endocrinology, 1995,136(1)’24-131.
    [135] Kurland ES, Rosen CJ, Cosman F, et al. Insulin-like growth factor-I in men with idiopathic osteoporosis[J] J. Clin Endocrinol Metab, 1997,82:2799.
    [136] Klein BY, Shohami E, Reikhinshtein Y. Seru-mediated osteogenic effects of head injury on cultured rat marrow stromal cells. Calcif Tissue Int 1999 Sep:65(3):217-22.
    [137] Beeton C. A, Brooks R. A, Rushton N. Circulating levels of insulin-like growth factor-1 and insulin-like growth factor binding Protein-3in patients with severe head in jury. The Journal of Bone and Joint Surgery 2002,84-B(3):434-439.
    [138] TommoTsvkAzAk I. ToshI RO VSR, Effect of TGF-βon insuline-like growth factor-I autorine/paracrine axis in cultured rat articular chond rocytes [J]. Exp Cell Res, 1994,215:9.
    [139] Langdahl-BL, Kassem-M, Moller-MK, et al. The effects of IGF-Ⅰand IGF-Ⅱon proliferation and diffeention of human osteoblasts and interaction with growth hormone [J]. Eur-J-Clin -Invest, 1998,28(3):176-183.
    [140] Oxlund H, Mosekilde Li, Ottoft G.Reduced concentration of collagen reducible cross links in human trabecullar bone with respect to age and osteoporosis[J]. Bone, 1996,19:479-484.
    [141] Rehn M, Pihlajaniemi T. Alpha-l(ⅩⅧ), a collagen chain with frequent interruptions in the collagenous sequence, a distinct tissue distribution and homology with typeⅩⅤcollagen. Proc Natl Acad Sci USA 1994,91(10):4234-4238.
    [142] Garnero P, Sornay-rendu E, Hapuy MC, et al. Increased bone turnover in late postmenopausal women is a major determinant of osteoporosis[J]. J Bone Miner Res, 1996,11:337-349.
    [143] Cheng DT, Dicesare P, Benya PD, et al. The presence of intermolecular disulfide crosslinks in typeⅢcollagen. J Biol Chem, 1993,258(11)7774-7778.
    [144] Mayne R, Brewton RG. New members of the collagen superfamily. Curr Opin Cell Biol, 1993,5(5):883-890.
    [145] OxlundH, BarckmanM, AndreassenT, et al. Reduced concentration of collagen cross links are associated with reducedstrength of bone[J]. Bone, 1995,17:365s-371s.
    [146]杨志明,余希杰,黄富国,等.外源性I型胶原对人胚骨膜成骨胞生物学特性的影响.华西医科大学报, 2001:32(l):l.
    [147] Martin RB, Ishida J. The reletive effets of collagen fiber orientation,porosity, density and mineralization on bone strength. J biomech, 1989,22(5):419-426.
    [148] Hiltunen A, AroHT, Vuorio E. Regulation of extracellar matrix genes during fracture healing in mice.Clin Orthop, 1993,297:23-27.
    [149] Liu SH, Yang RS, Shaikh RA, et al. Collagen in tendon,ligament and bone healing. Clin Orthop, 1995,318:265-278.
    [150] Borque WT, Gross M, Hall BK. Expression of four growth factors during fracture repair. Int J Dev Biol, 1993,37(4):573-579.
    [151] Longobardi S, Keay N, Ehrnborg C, et al. Growth hormone(GH) effects on bone and collagen turnover in healthy adults and its potential as a marker of GH abuse in sports:A double blind, placebo-controlled study. The GH-2000 Study Group.J Clin Endocrinol Metab, 2000,85(4):1505-1512.
    [152] Wallace JD, Cuneo RC, Lundberg PA, et al. Responses of markers of bone and collagenturnover to exercise,growth hormo ne(GH) administration and GH withdrawal in trained adult males. J Clin Endocrinol Metab, 2000,85(1):124-133.
    [153] Bostrom MPC, Salch KJ. Einhorn TA. Osteoinductive growth facors in preclinical fracture and long bone defects models. Orthop Clin of North AM, 1999,30(4):647
    [154] Hon g Z-k, Qiu X-Y, Sun J-R.Grafting polymerization of L-lactide on the surface of hydroxyapatite nano-crystals.Polymer, Polymer 45(2004):6699-6706.
    [155]邵英.聚L-乳酸/聚L-乳酸接枝的纳米羟基磷灰石复合材料(PLLA/PLLA-gHA)的生物相容性研究[D].吉林:吉林大学第一临床医院,2007.
    [156]彭威海.新型可吸收材料PLLA/PLLA-gHA的生物降解性及生物力学的实验研究[D].吉林:吉林大学第一临床医院,2009.
    [157] Leenslag J, Pennings A, Bos R,et al. Resorbable materials of poly(L-lactide). VII. In-vivo and vitro degradation. Biomate-rials. 1987,8(7):311.
    [158] Bostman O, Pihlajamaki H. Clinical biocompatibility of biodegradable orthopaedic implants for internal fixation. Bio-materials, 2000,21(24):2615-2621.
    [159] Makela A, Vainionpaa S, Vithonen K, et al. The effects of a penetrating biodrgradable implant on the growth plate. Clin Orthop Rel Res, 1989,241:300.
    [160] Hayden JM, Mohan S, Baylink DJ. The insulin-like growth factor system and the coupling of formation to resorption.Bone. 1995,17:93S-98S.
    [161] Scandberg M, Aro H, Multimaki P, et al. In situ localization of collagen production by chondrocytes and osteoblasts in fracture callus. J Bone Joint Surg(Am), 1989,71(1):69-77.
    [162] Vauhkonen M, Peltonen J, Karaharju E, et al. Collagen synthesis and mineralization in the early phase of distraction bone healing. BoneMiner, 1990,10(3):171- 181.