羊膜上皮细胞与化学去细胞肌肉支架联合移植治疗大鼠脊髓损伤的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
去细胞肌肉组织工程支架可作为生物工程支架支持神经元轴突再生。研究表明羊膜上皮细胞可分泌多种神经营养因子,促进神经元轴突的生长,是治疗神经系统疾病良好的种子细胞。本实验将化学去细胞肌肉做为支架材料,以大鼠羊膜上皮细胞做为种子细胞,共同移植入大鼠脊髓半横断损伤处,观察它们对脊髓损伤的治疗效果。
     本研究通过冻融和化学萃取方法制备了两种去细胞肌肉组织工程支架,并把它们分别植入大鼠半横断脊髓缺损处,4W后观察两种去细胞肌肉中的轴突再生情况。与冻融去细胞肌肉相比,化学去细胞肌肉可以更好的促进损伤脊髓的轴突再生,支架内再生轴突定向分布。化学去细胞肌肉支架移植不引起明显的炎症反应,其内血管再生良好,对胶质瘢痕的形成有一定抑制作用,化学去细胞肌肉与脊髓具有良好的生物相容性。羊膜上皮细胞与化学去细胞肌肉共同移植入大鼠脊髓(联合移植)后,与单纯化学去细胞肌肉支架移植比较,促进了轴突的再生,并在一定程度上促进了5-HT能神经纤维的再生,更重要的是,联合移植明显的促进了髓鞘的生成,通过BBB评分和神经电生理方法检测发现,联合移植更好的促进了大鼠患肢的功能恢复。
     总之,本研究证明化学去细胞肌肉组织工程支架可有效促进损伤脊髓的轴突再生,具有良好的生物相容性;并成功的利用羊膜上皮细胞与支架的共移植治疗大鼠的脊髓损伤,为脊髓损伤的组织工程治疗提供了理论和实验依据。
Spinal Cord Injury (SCI) is a serious debilitating illness resulting from transport, mining accidents and sporting accidents. It is reported that China currently has about 400 000 patients with SCI with an annual increase of about 1 million. The people afflicted with SCI are a big burden to both the society and the family. The pathophysiology of spinal cord injury is characterized by the primary injury followed by secondary injury processes that cause spinal cord hemorrhage, edema, axonal and neuronal necrosis, cavity formation, infarction and subsequent prolonged demyelination, and ultimately lead to the loss of motor and sensory function of spinal cord. At present, the treatment of spinal cord injury, on the one hand, is to prevent secondary pathological changes caused by mechanical injury from aggravating spinal cord damage, and to protect the injured spinal cord. On the other hand the treatment is to promote axonal regeneration and neural pathway reconstruction of the damaged area, to obtain functional recovery of damaged spinal cord. Spinal cord protection strategies can not promote axonal regeneration, but only minimize the destruction of axons and neurons caused by secondary damage. So the promotion of axon regeneration is the key to promote the rehabilitation of damaged spinal cord function in the treatment of spinal cord injury.
     The reasons for the mammalian central nervous system unable to regenerate include:①poor regenerative capacity of adult mammalian central nervous system②astrocytes stimulated by the immune response and excitotoxity surround the injured area, creating the glial scar which is the obstacle of axonal regeneration and neural pathway reconstruction.③inhibitors such as Nogo, MAG and proteoglycans to axonal regeneration within the injured spinal cord. Recent studies show that the neurotrophic factors, cell transplantation, tissue engineering, gene therapy and other means can promote axonal regeneration after spinal cord injury which improve the injured tissue microenvironment. The studies also shows that tissue engineering scaffolds in spinal cord injury not only can reduce the glial scar formation and guide axonal regeneration by forming a bridge structure, but also become an important aid tool for cell transplantation. For example, treatments for spinal cord injury by neurotrophic factors and cell transplantation are difficult to overcome the glial scar, but the neural tissue engineering approaches have acquired certain effect. Especially in the case when the primary injury results into partial fracture of spinal cord, nerve tissue engineering scaffolds can provide support and guidance for the regeneration of axons through the gap. Currently, nerve tissue engineering has made great development, but there is no one which can be used in clinical applications. Studies have shown that peripheral nerves can promote axonal regeneration of
     injured spinal cord. However, the weaknesses of peripheral nerve limit its further application. In the course of neural transplantation, the study found that extracellular matrix (laminin, fibronectin, heparan sulphate) of neural basement membrane plays an important role in axon regeneration. The skeletal muscle basement membrane is very similar to neural basement membrane, resulting the possibility of using muscle tissue to replace nerve as nerve tissue engineering scaffold. However, the transplantation of fresh skeletal muscle may produce a strong immune rejection. This problem can be solved by decellularization technology which can remove the skeletal muscle cells, effectively eliminating the antigenicity of skeletal muscle as a graft, and keep basement membrane components, making the transplant possible. Indeed, tissue engineering studies have proven that acellular muscle can replace nerve to promote axonal regeneration of injured peripheral nerve. Acellular muscles as nerve tissue engineering scaffolds have many advantages. First of all, the extracellular matrix components of acellular muscle have an important role in cell migration, adhesion, growth and metabolism. Second, the arrangement of acellular muscle structure is similar to that of neural tube, which provides sufficient space to support axonal growth, and is very important for the induction of neuronal regeneration. In addition acellular muscle eliminate cellular components and reduce the immunogenicity, possibly allowing allograft or xenograft. So acellular muscle may be the ideal tissue engineering scaffold for the treatment of spinal cord injury.
     The pathological process of spinal cord injury is very complicated. So the effect of a single treatment is often limited. At present, one common strategy of the nerve tissue engineering is to combine the tissue engineering scaffold and cell transplantation for treatment of spinal cord injury. The choice of seed cells is an important aspect of tissue engineering. Several factors must be considered, such as the role of neurotrophic factors released by seed cells, access ways, ethical issues and etc. Amniotic epithelial cells can secrete BDNF, NT-3 and NGF and other neurotrophic factors. After spinal cord injury ,the application of NT-3, BDNF and NGF not only reduce the edema and the blood- spinal cord barrier injury, providing a protective effect on neurons, but also can promote the regeneration of injured spinal cord axons and be beneficial to functional recovery. The study of amniotic epithelial cell transplantation in rat spinal cord contusion injury model found that amniotic epithelial cell transplantation promote axonal regeneration of injured spinal cord and recovery of hind limb function of the animals. In addition, the amniotic membrane is a product derived from fetal which is exposure to maternal immune system surveillance. The cell surface has low human leucocyte antigen-DR expression and no expression of HLA-A, B, C antigens. So its immunogenicity is very low. Amniotic epithelial cells also secrete a variety of soluble molecules which can reduce the specific and non-specific immunological activity of immune cells and play an anti-inflammatory role. And the transplantation of amnion cells did not present the risk of tumorigenicity. Amniotic membrane is a part of postpartum waste tissue. So it has plenty source and no ethics problem. Therefore, amniotic epithelial cells may be the ideal seed cells for spinal cord repair.
     This study was to fabricate chemically extracted acellular muscle and freeze-thawed acellular muscle scaffolds, using them to treat hemisected adult rat spinal cord, and comparing their roles in promoting axon regeneration. After going through screening, chemically extracted acellular muscle scaffolds and amniotic epithelial cells were co-transplanted into the hemisected adult rat spinal cord, to observe the effects of treatment. Details are as follows.
     1. The fabrication of acellular muscle scaffolds, observation of scaffold’s histological structure, and identification of scaffold’s composition
     The acellular muscle scaffolds were prepared by SDS and TritonX-100 extraction or freeze-thaw method. Conventional HE staining and Weigert-Van Gieson staining were used to observe the scaffold’s histological structure, and to identify the scaffold’s composition. The dry-wet weight method was used to detect water content of scaffolds. Image-Pro Plus 6 image analysis software was used to measure the diameter of scaffold’s pipeline. HE staining showed that chemically extracted acellular muscle scaffold completely removes the cell components and retain the fiber components of extracellular matrix, with the spatial configuration of fiber components being the pipeline structure parallel with the muscle, suggesting that the chemically extracted acellular muscle scaffold was successfully made. HE staining showed that in freeze-thawed acellular muscle, nucleus and stripes of cytoplasm disappeared, suggesting that cell structures were damaged and freeze-thaw method also removed living cells. It also showed that freeze-thawed scaffold contained a large number of residual muscle cell components. Between them were the parallel pipeline structures, suggesting that freeze-thawed acellular muscle scaffolds were prepared successfully. Weigert-Van Gieson staining showed that the chemically extracted acellular muscle scaffold was mainly composed of collagen and elastic fibers and that the freeze-thawed scaffold, apart from the above components, also included components of residual myocytes. The dry-wet weight measurements showed that the two scaffolds contained large amounts of water, which is helpful for the transport of oxygen and nutrients. Image analysis showed that the diameters of pipeline in two kinds of scaffolds are about 100 micrometers, which will help the migration and adhesion of cells.
     2. Screening of the acellular muscle scaffolds
     Chemically extracted acellular muscle and freeze-thawed acellular muscle scaffolds were respectively implanted into the hemisected adult rat spinal cord. At 7d, 14d and 28d after transplantation, ED-1 immunohistochemistry showed, both compared with the injured group, chemically extracted acellular muscle and freeze-thawed acellular muscle scaffolds don't cause significant foreign body rejection. At 28d after transplantation, Holmes silver staining revealed that the chemically extracted acellular muscle can better promote axonal regeneration after spinal cord injury, with regenerated axons in a strikingly organized and linear fashion.
     3. The biocompatibility of chemically extracted acellular muscle grafts as biomatrices in experimental spinal cord injury in rats
     We further verified the biocompatibility of chemically extracted acellular muscle scaffold. At 28d after transplantation, alkaline phosphatase staining showed that angiogenesis is good in chemically extracted acellular muscle scaffold. GFAP immunohistochemistry showed that the distribution of astrocytes around the chemically extracted acellular muscle scaffold changed in some areas, suggesting the formation of the glial scar is inhibited. HRP neural tracing results showed that chemically extracted acellular muscle scaffold can promote the regeneration of nerve fibers in the spinal cord itself. Therefore, the chemically extracted acellular muscle scaffold has good biocompatibility with rat spinal cord.
     4. Chemically extracted acellular muscle scaffold seeded with amniotic epithelial cells to promote spinal cord repair
     Amniotic epithelial cells and chemically extracted acellular muscle grafts were co-transplanted into the hemisected adult rat spinal cord. At 28d after transplantation, the immunofluorescence staining found, in comparison with pure scaffold transplantation, co-transplantation promoted the regeneration of nerve axons, and to some extent, facilitated the 5-HT nerve fiber regeneration and, more importantly, significantly promoted the myelination of nerve fibers. The BBB tests and nerve electrophysiological methods also detected the cotransplantation promote functional recovery of rat limb better.
     In conclusion, this study showed the immune response after chemically extracted acellular muscle scaffold transplantation is weak. The chemically extracted acellular muscle scaffolds possess good biocompatibility and can promote robust regrowth of axons in spinal cord with regenerating axons in a strikingly organized and linear fashion. This study also successfully used co-transplantation of amniotic epithelial cells and chemically extracted acellular muscle scaffolds to treat spinal cord injury, providing a theoretical and experimental basis for the tissue engineering for spinal cord injury.
引文
[1]蒋晖,金大地,江建明.神经干细胞在脊髓损伤修复中的应用研究进展[J].第一军医大学学报, 2001,21: 114-116.
    [2] Taylor SJ, Rosenzweig ES, McDonald JW, 3rd, et al. Delivery of neurotrophin-3 from fibrin enhances neuronal fiber sprouting after spinal cord injury[J]. J Control Release, 2006,113:226-235.
    [3] Sharma HS. A select combination of neurotrophins enhances neuroprotection and functional recovery following spinal cord injury[J]. Ann N Y Acad Sci, 2007,1122:95-111.
    [4] Song XY, Li F, Zhang FH, et al. Peripherally-derived BDNF promotes regeneration of ascending sensory neurons after spinal cord injury[J]. PLoS ONE, 2008,3:e1707.
    [5] Pan HC, Cheng FC, Lai SZ, et al. Enhanced regeneration in spinal cord injury by concomitant treatment with granulocyte colony-stimulating factor and neuronal stem cells[J]. J Clin Neurosci, 2008,15:656-664.
    [6] Novikova LN, Pettersson J, Brohlin M, et al. Biodegradable poly-beta- hydroxybutyrate scaffold seeded with Schwann cells to promote spinal cord repair[J]. Biomaterials, 2008,29:1198-1206.
    [7] Novikova LN, Novikov LN, Kellerth JO. Biopolymers and biodegradable smart implants for tissue regeneration after spinal cord injury[J]. Curr Opin Neurol, 2003,16:711-715.
    [8] Meng XT, Li C, Dong ZY, et al. Co-transplantation of bFGF-expressing amniotic epithelial cells and neural stem cells promotes functional recovery in spinal cord-injured rats[J]. Cell Biol Int, 2008,32:1546-1558.
    [9] Bresnahan JC, King JS, Martin GF, et al. A neuroanatomical analysis of spinal cord injury in the rhesus monkey (Macaca mulatta) [J]. J Neurol Sci, 1976,28:521-542.
    [10] Blight AR. Axonal physiology of chronic spinal cord injury in the cat:intracellular recording in vitro[J]. Neuroscience, 1983,10:1471-1486.
    [11] Kakulas BA. Pathology of spinal injuries[J]. Cent Nerv Syst Trauma, 1984,1:117-129.
    [12] Wallace MC, Tator CH, Lewis AJ. Chronic regenerative changes in the spinal cord after cord compression injury in rats[J]. Surg Neurol, 1987,27:209-219.
    [13] Sandler AN, Tator CH. Review of the effect of spinal cord trama on the vessels and blood flow in the spinal cord[J]. J Neurosurg, 1976,45:638-646.
    [14] Collins WF. A review and update of experiment and clinical studies of spinal cord injury[J]. Paraplegia, 1983,21:204-219.
    [15] Hall ED, Yonkers PA, Horan KL, et al. Correlation between attenuation of posttraumatic spinal cord ischemia and preservation of tissue vitamin E by the 21-aminosteroid U74006F: evidence for an in vivo antioxidant mechanism[J]. J Neurotrauma, 1989,6:169-176.
    [16] Tator CH, Fehlings MG. Review of the secondary injury theory of acute spinal cord trauma with emphasis on vascular mechanisms [ J] . J Neurosurg, 1991,75:15-26.
    [17] Allen AR. SURGERY OF EXPERIMENTAL LESION OF SPINAL CORD EQUIVALENT TO CRUSH INJURY OF FRACTURE DISLOCATION OF SPINAL COLUMN[J]. jama-journal of the american medical association, 1911,57:878-880.
    [18] Allen AR. Remarks on the Histopathological Changes in the Spinal Cord Due To Impact. An Experimental Study[J]. The Journal of Nervous and Mental Disease, 1914, 41 141-147.
    [19] Ducker TB, Kindt GW, Kempf LG. Pathological findings in acute experimental spinal cord trauma[J]. J Neurosurg, 1971,35:700-708.
    [20] Nemecek S. Morphological evidence of microcirculatory disturbances in experimental spinal cord trauma[J]. Adv Neurol, 1978,20:395-405.
    [21] Tator CH. Experimental and clinical studies of the pathophysiology and management of acute spinal cord injury [ J ] . J Spinal Cord Med,1996,19:206-214.
    [22] Guha A, Tator CH. Acute cardiovascular effects of experimental spinal cord injury[J]. J Trauma, 1988,28:481-490.
    [23] Akdemir H, Pasaoglu A, Ozturk F, et al. Histopathology of experimental spinal cord trauma. Comparison of treatment with TRH, naloxone, and dexamethasone[J]. Res Exp Med (Berl), 1992,192:177-183.
    [24] Doble A. The role of excitotoxicity in neurodegenerative disease: implications for therapy[J]. Pharmacol Ther, 1999,81:163-221.
    [25] Beattie MS. Inflammation and apoptosis: linked therapeutic targets in spinal cord injury[J]. Trends Mol Med, 2004,10:580-583.
    [26] Faden AI, Halt P. Platelet-activating factor reduces spinal cord blood flow and causes behavioral deficits after intrathecal administration in rats through a specific receptor mechanism[J]. J Pharmacol Exp Ther, 1992,261:1064-1070.
    [27] Emerich DF, Dean RL, 3rd, Bartus RT. The role of leukocytes following cerebral ischemia: pathogenic variable or bystander reaction to emerging infarct? [J] Exp Neurol, 2002,173:168-181.
    [28] Dirnagl U, Iadecola C, Moskowitz MA. Pathobiology of ischaemic stroke: an integrated view[J]. Trends Neurosci, 1999,22:391-397.
    [29] Taoka Y, Okajima K. Spinal cord injury in the rat[J]. Prog Neurobiol, 1998,56:341-358.
    [30] Young W. Role of calcium in central nervous system injuries[J]. J Neurotrauma, 1992,9 Suppl 1:S9-25.
    [31] Fawcett JW, Asher RA. The glial scar and central nervous system repair[J]. Brain Res Bull, 1999,49:377-391.
    [32] Maier IC, Schwab ME. Sprouting, regeneration and circuit formation in the injured spinal cord: factors and activity[J]. Philos Trans R Soc Lond B Biol Sci, 2006,361:1611-1634.
    [33] Taketomi M, Kinoshita N, Kimura K, et al. Nogo-A expression in mature oligodendrocytes of rat spinal cord in association with specific molecules[J].Neurosci Lett, 2002,332:37-40.
    [34] Reynolds BA, Weiss S. Generation of neurons and astrocytes from isolated cells of the adult mammalian central nervous system [ J ] . Science, 1992,255:1707-1710.
    [35] Gage FH, Coates PW, Palmer TD, et al. Survival and differentiation of adult neuronal progenitor cells transplanted to the adult brain[J]. Proc Natl Acad Sci U S A, 1995,92:11879-11883.
    [36] Shihabuddin LS, Horner PJ, Ray J, et al. Adult spinal cord stem cells generate neurons after transplantation in the adult dentate gyrus[J]. J Neurosci, 2000,20:8727-8735.
    [37] Martino G, Pluchino S. The therapeutic potential of neural stem cell[sJ]. Nat Rev Neurosci, 2006,7:395-406.
    [38] Gage FH, Verma IM. Stem cells at the dawn of the 21st century[J]. Proc Natl Acad Sci U S A, 2003,100 Suppl 1:11817-11818.
    [39] Keirstead HS, Nistor G, Bernal G, et al. Human embryonic stem cell-derived oligodendrocyte progenitor cell transplants remyelinate and restore locomotion after spinal cord injury[J]. J Neurosci, 2005,25:4694-4705.
    [40] Cao Q, Benton RL, Whittemore SR. Stem cell repair of central nervous system injury[J]. J Neurosci Res, 2002,68:501-510.
    [41] Fischbach GD, Fischbach RL. Stem cells: science, policy, and ethics[J]. J Clin Invest, 2004,114:1364-1370.
    [42] Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells[J]. Science, 1999,284:143-147.
    [43] Satake K, Lou J, Lenke LG. Migration of mesenchymal stem cells through cerebrospinal fluid into injured spinal cord tissue[J]. Spine (Phila Pa 1976), 2004,29:1971-1979.
    [44] Hall S. Axonal regeneration through acellular muscle grafts[J]. J Anat, 1997,190 ( Pt 1):57-71.
    [45] Oudega M, Xu XM. Schwann cell transplantation for repair of the adult spinalcord[J]. J Neurotrauma, 2006,23:453-467.
    [46] Lu J, Feron F, Ho SM, et al. Transplantation of nasal olfactory tissue promotes partial recovery in paraplegic adult rats[J]. Brain Res, 2001,889:344-357.
    [47] Shen H, Tang Y, Wu Y, et al. Influences of olfactory ensheathing cells transplantation on axonal regeneration in spinal cord of adult rats[J]. Chin J Traumatol, 2002,5:136-141.
    [48] Boulenguez P, Vinay L. Strategies to restore motor functions after spinal cord injury[J]. Curr Opin Neurobiol, 2009,19:587-600.
    [49] Andrews MR, Stelzner DJ. Evaluation of olfactory ensheathing and schwann cells after implantation into a dorsal injury of adult rat spinal cord[J]. J Neurotrauma, 2007,24:1773-1792.
    [50] Miki T, Lehmann T, Cai H, et al. Stem cell characteristics of amniotic epithelial cells[J]. Stem Cells, 2005,23:1549-1559.
    [51] Takashima S, Ise H, Zhao P, et al. Human amniotic epithelial cells possess hepatocyte-like characteristics and functions [ J ] . Cell Struct Funct, 2004,29:73-84.
    [52] Ilancheran S, Michalska A, Peh G, et al. Stem cells derived from human fetal membranes display multilineage differentiation potential[J]. Biol Reprod, 2007,77:577-588.
    [53] Uchida S, Inanaga Y, Kobayashi M, et al. Neurotrophic function of conditioned medium from human amniotic epithelial cells [ J ] . J Neurosci Res, 2000,62:585-590.
    [54] Sakuragawa N, Tohyama J, Yamamoto H. Immunostaining of human amniotic epithelial cells: possible use as a transgene carrier in gene therapy for inborn errors of metabolism[J]. Cell Transplant, 1995,4:343-346.
    [55] Akle CA, Adinolfi M, Welsh KI, et al. Immunogenicity of human amniotic epithelial cells after transplantation into volunteers [ J ] . Lancet, 1981,2:1003-1005.
    [56] Toda A, Okabe M, Yoshida T, et al. The potential of amnioticmembrane/amnion-derived cells for regeneration of various tissues[J]. J Pharmacol Sci, 2007,105:215-228.
    [57] Sankar V, Muthusamy R. Role of human amniotic epithelial cell transplantation in spinal cord injury repair research[J]. Neuroscience, 2003,118:11-17.
    [58] Wu ZY, Hui GZ, Lu Y, et al. Transplantation of human amniotic epithelial cells improves hindlimb function in rats with spinal cord injury[J]. Chin Med J (Engl), 2006,119:2101-2107.
    [59] Nomura H, Tator CH, Shoichet MS. Bioengineered strategies for spinal cord repair[J]. J Neurotrauma, 2006,23:496-507.
    [60] Mackinnon SE, Dellon AL. Clinical nerve reconstruction with a bioabsorbable polyglycolic acid tube[J]. Plast Reconstr Surg, 1990,85:419-424.
    [61] Matsumoto K, Ohnishi K, Kiyotani T, et al. Peripheral nerve regeneration across an 80-mm gap bridged by a polyglycolic acid (PGA)-collagen tube filled with laminin-coated collagen fibers: a histological and electrophysiological evaluation of regenerated nerves[J]. Brain Res, 2000,868:315-328.
    [62] Patist CM, Mulder MB, Gautier SE, et al. Freeze-dried poly(D,L-lactic acid) macroporous guidance scaffolds impregnated with brain-derived neurotrophic factor in the transected adult rat thoracic spinal cord[J]. Biomaterials, 2004,25:1569-1582.
    [63] Athanasiou KA, Niederauer GG, Agrawal CM. Sterilization, toxicity, biocompatibility and clinical applications of polylactic acid/polyglycolic acid copolymers[J]. Biomaterials, 1996,17:93-102.
    [64] Gunatillake PA, Adhikari R. Biodegradable synthetic polymers for tissue engineering[J]. Eur Cell Mater, 2003,5:1-16; discussion 16.
    [65] Teng YD, Lavik EB, Qu X, et al. Functional recovery following traumatic spinal cord injury mediated by a unique polymer scaffold seeded with neural stem cells[J]. Proc Natl Acad Sci U S A, 2002,99:3024-3029.
    [66] Olson HE, Rooney GE, Gross L, et al. Neural stem cell- and Schwann cell-loaded biodegradable polymer scaffolds support axonal regeneration in thetransected spinal cord[J]. Tissue Eng Part A, 2009,15:1797-1805.
    [67] Nisbet DR, Pattanawong S, Ritchie NE, et al. Interaction of embryonic cortical neurons on nanofibrous scaffolds for neural tissue engineering[J]. J Neural Eng, 2007,4:35-41.
    [68] Gautier SE, Oudega M, Fragoso M, et al. Poly(alpha-hydroxyacids) for application in the spinal cord: resorbability and biocompatibility with adult rat Schwann cells and spinal cord[J]. J Biomed Mater Res, 1998,42:642-654.
    [69] Midha R, Munro CA, Dalton PD, et al. Growth factor enhancement of peripheral nerve regeneration through a novel synthetic hydrogel tube[J]. J Neurosurg, 2003,99:555-565.
    [70] Katayama Y, Montenegro R, Freier T, et al. Coil-reinforced hydrogel tubes promote nerve regeneration equivalent to that of nerve autografts [ J] . Biomaterials, 2006,27:505-518.
    [71] Tsai EC, Dalton PD, Shoichet MS, et al. Matrix inclusion within synthetic hydrogel guidance channels improves specific supraspinal and local axonal regeneration after complete spinal cord transection [ J ] . Biomaterials, 2006,27:519-533.
    [72] Giannetti S, Lauretti L, Fernandez E, et al. Acrylic hydrogel implants after spinal cord lesion in the adult rat[J]. Neurol Res, 2001,23:405-409.
    [73] Tsai EC, Dalton PD, Shoichet MS, et al. Synthetic hydrogel guidance channels facilitate regeneration of adult rat brainstem motor axons after complete spinal cord transection[J]. J Neurotrauma, 2004,21:789-804.
    [74] Belkas JS, Munro CA, Shoichet MS, et al. Long-term in vivo biomechanical properties and biocompatibility of poly(2-hydroxyethyl methacrylate-co-methyl methacrylate) nerve conduits[J]. Biomaterials, 2005,26:1741-1749.
    [75] Parker AL, Fisher KD, Oupicky D, et al. Enhanced gene transfer activity of peptide-targeted gene-delivery vectors[J]. J Drug Target, 2005,13:39-51.
    [76] Prokopova-Kubinova S, Vargova L, Tao L, et al. Poly[N-(2- hydroxypropyl) methacrylamide] polymers diffuse in brain extracellular space with sametortuosity as small molecules[J]. Biophys J, 2001,80:542-548.
    [77] Woerly S, Pinet E, De Robertis L, et al. Heterogeneous PHPMA hydrogels for tissue repair and axonal regeneration in the injured spinal cord[J]. J Biomater Sci Polym Ed, 1998,9:681-711.
    [78] Woerly S, Doan VD, Evans-Martin F, et al. Spinal cord reconstruction using NeuroGel implants and functional recovery after chronic injury[J]. J Neurosci Res, 2001,66:1187-1197.
    [79] Woerly S, Doan VD, Sosa N, et al. Prevention of gliotic scar formation by NeuroGel allows partial endogenous repair of transected cat spinal cord[J]. J Neurosci Res, 2004,75:262-272.
    [80] Loh NK, Woerly S, Bunt SM, et al. The regrowth of axons within tissue defects in the CNS is promoted by implanted hydrogel matrices that contain BDNF and CNTF producing fibroblasts[J]. Exp Neurol, 2001,170:72-84.
    [81] Shoichet MS, Rein DH. In vivo biostability of a polymeric hollow fibre membrane for cell encapsulation[J]. Biomaterials, 1996,17:285-290.
    [82] Flanagan LA, Ju YE, Marg B, et al. Neurite branching on deformable substrates[J]. Neuroreport, 2002,13:2411-2415.
    [83] Aebischer P, Guenard V, Valentini RF. The morphology of regenerating peripheral nerves is modulated by the surface microgeometry of polymeric guidance channels[J]. Brain Res, 1990,531:211-218.
    [84] Xu XM, Chen A, Guenard V, et al. Bridging Schwann cell transplants promote axonal regeneration from both the rostral and caudal stumps of transected adult rat spinal cord[J]. J Neurocytol, 1997,26:1-16.
    [85] Bamber NI, Li H, Lu X, et al. Neurotrophins BDNF and NT-3 promote axonal re-entry into the distal host spinal cord through Schwann cell-seeded mini-channels[J]. Eur J Neurosci, 2001,13:257-268.
    [86] Fouad K, Schnell L, Bunge MB, et al. Combining Schwann cell bridges and olfactory-ensheathing glia grafts with chondroitinase promotes locomotor recovery after complete transection of the spinal cord [J]. J Neurosci,2005,25:1169-1178.
    [87] Iannotti C, Li H, Yan P, et al. Glial cell line-derived neurotrophic factor-enriched bridging transplants promote propriospinal axonal regeneration and enhance myelination after spinal cord injury[J]. Exp Neurol, 2003,183:379-393.
    [88] Alcantar NA, Aydil ES, Israelachvili JN. Polyethylene glycol-coated biocompatible surfaces[J]. J Biomed Mater Res, 2000,51:343-351.
    [89] Luo J, Borgens R, Shi R. Polyethylene glycol immediately repairs neuronal membranes and inhibits free radical production after acute spinal cord injury[J]. J Neurochem, 2002,83:471-480.
    [90] Luo J, Shi R. Diffusive oxidative stress following acute spinal cord injury in guinea pigs and its inhibition by polyethylene glycol[J]. Neurosci Lett, 2004,359:167-170.
    [91] Luo J, Shi R. Polyethylene glycol inhibits apoptotic cell death following traumatic spinal cord injury[J]. Brain Res, 2007,1155:10-16.
    [92] Borgens RB, Shi R, Bohnert D. Behavioral recovery from spinal cord injury following delayed application of polyethylene glycol [ J ] . J Exp Biol, 2002,205:1-12.
    [93] During MJ, Freese A, Sabel BA, et al. Controlled release of dopamine from a polymeric brain implant: in vivo characterization [ J ] . Ann Neurol, 1989,25:351-356.
    [94] Freese A, Sabel BA, Saltzman WM, et al. Controlled release of dopamine from a polymeric brain implant: in vitro characterization [ J ] . Exp Neurol, 1989,103:234-238.
    [95] Krewson CE, Klarman ML, Saltzman WM. Distribution of nerve growth factor following direct delivery to brain interstitium[J]. Brain Res, 1995,680:196-206.
    [96] Krewson CE, Saltzman WM. Transport and elimination of recombinant human NGF during long-term delivery to the brain[J]. Brain Res, 1996,727:169-181.
    [97] Powell EM, Sobarzo MR, Saltzman WM. Controlled release of nerve growth factor from a polymeric implant[J]. Brain Res, 1990,515:309-311.
    [98] Barras FM, Pasche P, Bouche N, et al. Glial cell line-derived neurotrophic factor released by synthetic guidance channels promotes facial nerve regeneration in the rat[J]. J Neurosci Res, 2002,70:746-755.
    [99] Schepsis AA, Greenleaf J. Prosthetic materials for anterior cruciate ligament reconstruction[J]. Orthop Rev, 1990,19:984-991.
    [100] Khan T, Dauzvardis M, Sayers S. Carbon filament implants promote axonal growth across the transected rat spinal cord[J]. Brain Res, 1991,541:139-145.
    [101] Chauhan NB, Figlewicz HM, Khan T. Carbon filaments direct the growth of postlesional plastic axons after spinal cord injury[J]. Int J Dev Neurosci, 1999,17:255-264.
    [102] Liu LS, Khan T, Sayers ST, et al. Electrophysiological improvement after co-implantation of carbon filaments and fetal tissue in the contused rat spinal cord[J]. Neurosci Lett, 1995,200:199-202.
    [103] McKenzie JL, Waid MC, Shi R, et al. Decreased functions of astrocytes on carbon nanofiber materials[J]. Biomaterials, 2004,25:1309-1317.
    [104] Stokols S, Tuszynski MH. The fabrication and characterization of linearly oriented nerve guidance scaffolds for spinal cord injury[J]. Biomaterials, 2004,25:5839-5846.
    [105] Stokols S, Tuszynski MH. Freeze-dried agarose scaffolds with uniaxial channels stimulate and guide linear axonal growth following spinal cord injury[J]. Biomaterials, 2006,27:443-451.
    [106] Kataoka K, Suzuki Y, Kitada M, et al. Alginate enhances elongation of early regenerating axons in spinal cord of young rats [ J ] . Tissue Eng, 2004,10:493-504.
    [107] Suzuki Y, Kitaura M, Wu S, et al. Electrophysiological and horseradish peroxidase-tracing studies of nerve regeneration through alginate-filled gap in adult rat spinal cord[J]. Neurosci Lett, 2002,318:121-124.
    [108] Wu S, Suzuki Y, Kitada M, et al. Migration, integration, and differentiation of hippocampus-derived neurosphere cells after transplantation into injured ratspinal cord[J]. Neurosci Lett, 2001,312:173-176.
    [109] Prang P, Muller R, Eljaouhari A, et al. The promotion of oriented axonal regrowth in the injured spinal cord by alginate-based anisotropic capillary hydrogels[J]. Biomaterials, 2006,27:3560-3569.
    [110] Zimmermann U, Thurmer F, Jork A, et al. A novel class of amitogenic alginate microcapsules for long-term immunoisolated transplantation[J]. Ann N Y Acad Sci, 2001,944:199-215.
    [111] Stabenfeldt SE, Garcia AJ, LaPlaca MC. Thermoreversible laminin- functionalized hydrogel for neural tissue engineering[J]. J Biomed Mater Res A, 2006,77:718-725.
    [112] Zhong Y, Bellamkonda RV. Controlled release of anti-inflammatory agent alpha-MSH from neural implants[J]. J Control Release, 2005,106:309-318.
    [113] Itoh S, Suzuki M, Yamaguchi I, et al. Development of a nerve scaffold using a tendon chitosan tube[J]. Artif Organs, 2003,27:1079-1088.
    [114] Yuan Y, Zhang P, Yang Y, et al. The interaction of Schwann cells with chitosan membranes and fibers in vitro[J]. Biomaterials, 2004,25:4273-4278.
    [115] Willerth SM, Sakiyama-Elbert SE. Approaches to neural tissue engineering using scaffolds for drug delivery[J]. Adv Drug Deliv Rev, 2007,59:325-338.
    [116] Cheng H, Huang YC, Chang PT, et al. Laminin-incorporated nerve conduits made by plasma treatment for repairing spinal cord injury[J]. Biochem Biophys Res Commun, 2007,357:938-944.
    [117] Gamez E, Goto Y, Nagata K, et al. Photofabricated gelatin-based nerve conduits: nerve tissue regeneration potentials[J]. Cell Transplant, 2004,13:549-564.
    [118] Mahoney MJ, Krewson C, Miller J, et al. Impact of cell type and density on nerve growth factor distribution and bioactivity in 3-dimensional collagen gel cultures[J]. Tissue Eng, 2006,12:1915-1927.
    [119] Wissink MJ, Beernink R, Pieper JS, et al. Immobilization of heparin to EDC/NHS-crosslinked collagen. Characterization and in vitro evaluation[J]. Biomaterials, 2001,22:151-163.
    [120] Yoshii S, Oka M, Shima M, et al. Bridging a spinal cord defect using collagen filament[J]. Spine (Phila Pa 1976), 2003,28:2346-2351.
    [121] Watanabe K, Nakamura M, Okano H, et al. Establishment of three-dimensional culture of neural stem/progenitor cells in collagen Type-1 Gel[J]. Restor Neurol Neurosci, 2007,25:109-117.
    [122] Klaver CL, Caplan MR. Bioactive surface for neural electrodes: decreasing astrocyte proliferation via transforming growth factor-beta1[J]. J Biomed Mater Res A, 2007,81:1011-1016.
    [123] Levesque SG, Shoichet MS. Synthesis of cell-adhesive dextran hydrogels and macroporous scaffolds[J]. Biomaterials, 2006,27:5277-5285.
    [124] Herbert CB, Bittner GD, Hubbell JA. Effects of fibinolysis on neurite growth from dorsal root ganglia cultured in two- and three-dimensional fibrin gels[J]. J Comp Neurol, 1996,365:380-391.
    [125] Schense JC, Hubbell JA. Cross-linking exogenous bifunctional peptides into fibrin gels with factor XIIIa[J]. Bioconjug Chem, 1999,10:75-81.
    [126] Itosaka H, Kuroda S, Shichinohe H, et al. Fibrin matrix provides a suitable scaffold for bone marrow stromal cells transplanted into injured spinal cord: a novel material for CNS tissue engineering [ J ] . Neuropathology, 2009,29:248-257.
    [127] Ahmed Z, Underwood S, Brown RA. Nerve guide material made from fibronectin: assessment of in vitro properties[J]. Tissue Eng, 2003,9:219-231.
    [128] Ejim OS, Blunn GW, Brown RA. Production of artificial-orientated mats and strands from plasma fibronectin: a morphological study[J]. Biomaterials, 1993,14:743-748.
    [129] Brown RA, Blunn GW, Ejim OS. Preparation of orientated fibrous mats from fibronectin: composition and stability[J]. Biomaterials, 1994,15:457-464.
    [130] King VR, Henseler M, Brown RA, et al. Mats made from fibronectin support oriented growth of axons in the damaged spinal cord of the adult rat[J]. Exp Neurol, 2003,182:383-398.
    [131] King VR, Phillips JB, Brown RA, et al. The effects of treatment with antibodies to transforming growth factor beta1 and beta2 following spinal cord damage in the adult rat[J]. Neuroscience, 2004,126:173-183.
    [132] Wang KK, Nemeth IR, Seckel BR, et al. Hyaluronic acid enhances peripheral nerve regeneration in vivo[J]. Microsurgery, 1998,18:270-275.
    [133] Gupta D, Tator CH, Shoichet MS. Fast-gelling injectable blend of hyaluronan and methylcellulose for intrathecal, localized delivery to the injured spinal cord[J]. Biomaterials, 2006,27:2370-2379.
    [134] De Laporte L, Shea LD. Matrices and scaffolds for DNA delivery in tissue engineering[J]. Adv Drug Deliv Rev, 2007,59:292-307.
    [135] Madigan NN, McMahon S, O'Brien T, et al. Current tissue engineering and novel therapeutic approaches to axonal regeneration following spinal cord injury using polymer scaffolds[J]. Respir Physiol Neurobiol, 2009,169:183-199.
    [136] Krych AJ, Rooney GE, Chen B, et al. Relationship between scaffold channel diameter and number of regenerating axons in the transected rat spinal cord[J]. Acta Biomater, 2009,5:2551-2559.
    [137] Lacroix D, Chateau A, Ginebra MP, et al. Micro-finite element models of bone tissue-engineering scaffolds[J]. Biomaterials, 2006,27:5326-5334.
    [138] Sanders FK. THE REPAIR OF LARGE GAPS IN THE PERIPHERAL NERVES[J]. Brain 1942,65:281-337.
    [139] Mackinnon SE, Hudson AR, Falk RE, et al. Peripheral nerve allograft: an assessment of regeneration across pretreated nerve allografts[J]. Neurosurgery, 1984,15:690-693.
    [140] Gulati AK. Evaluation of acellular and cellular nerve grafts in repair of rat peripheral nerve[J]. J Neurosurg, 1988,68:117-123.
    [141] Borschel GH, Kia KF, Kuzon WM, Jr., et al. Mechanical properties of acellular peripheral nerve[J]. J Surg Res, 2003,114:133-139.
    [142] Wei HJ, Liang HC, Lee MH, et al. Construction of varying porous structures in acellular bovine pericardia as a tissue-engineering extracellular matrix[J].Biomaterials, 2005,26:1905-1913.
    [143] Grauss RW, Hazekamp MG, Oppenhuizen F, et al. Histological evaluation of decellularised porcine aortic valves: matrix changes due to different decellularisation methods[J]. Eur J Cardiothorac Surg, 2005,27:566-571.
    [144] Kallenbach K, Leyh RG, Lefik E, et al. Guided tissue regeneration: porcine matrix does not transmit PERV[J]. Biomaterials, 2004,25:3613-3620.
    [145] Islam S, Ogane K, Ohkuma H, et al. Usefulness of acellular dermal graft as a dural substitute in experimental model[J]. Surg Neurol, 2004,61:297-302; discussion 303.
    [146] Keynes RJ, Hopkins WG, Huang LH. Regeneration of mouse peripheral nerves in degenerating skeletal muscle: guidance by residual muscle fibre basement membrane[J]. Brain Res, 1984,295:275-281.
    [147] Glasby MA, Gschmeissner S, Hitchcock RJ, et al. Regeneration of the sciatic nerve in rats. The effect of muscle basement membrane[J]. J Bone Joint Surg Br, 1986,68:829-833.
    [148] Fansa H, Keilhoff G. Comparison of different biogenic matrices seeded with cultured Schwann cells for bridging peripheral nerve defects[J]. Neurol Res, 2004,26:167-173.
    [149] Meek MF, den Dunnen WF, Schakenraad JM, et al. Evaluation of several techniques to modify denatured muscle tissue to obtain a scaffold for peripheral nerve regeneration[J]. Biomaterials, 1999,20:401-408.
    [150] Cheng H, Cao Y, Olson L. Spinal cord repair in adult paraplegic rats: partial restoration of hind limb function[J]. Science, 1996,273:510-513.
    [151] Brown AL, Brook-Allred TT, Waddell JE, et al. Bladder acellular matrix as a substrate for studying in vitro bladder smooth muscle-urothelial cell interactions[J]. Biomaterials, 2005,26:529-543.
    [152]李培建,胥少汀.肌基膜管移植及神经生长因子对脊髓横断性损伤的修复作用[J].中国脊柱脊髓杂志, 2000,10:220-223.
    [153]刘维永,金振晓,顾春虎,赵东锷,谭红梅.去细胞猪主动脉瓣的构建及性能鉴定[J].中华医学杂志, 2005,85:324-327.
    [154]杨元,沙秋霖.山羊绒/绵羊毛混纺纱的定量分析[J].毛纺科技, 2008:43-46.
    [155]许扬赵英凯,毕明刚,刘妍. Image Pro Plus图像分析软件定量鸡胚尿囊膜血管新生面积的方法[J].中国比较医学杂志, 2007,17:745-748.
    [156]朱庆,刘艳霞,李刚,蔡敏,王敏伟.改良大鼠蛛网膜下腔出血模型[J].中国药理学通报, 2006,22:1144-1146.
    [157] Enomoto M, Wakabayashi Y, Qi ML, et al. Present situation and future aspects of spinal cord regeneration[J]. J Orthop Sci, 2004,9:108-112.
    [158] Nakamura M, Okano H, Toyama Y, et al. Transplantation of embryonic spinal cord-derived neurospheres support growth of supraspinal projections and functional recovery after spinal cord injury in the neonatal ra[tJ]. J Neurosci Res, 2005,81:457-468.
    [159] Houstava L, Dubovy P, Haninec P, et al. An alternative preparation of the acellular muscle graft for reconstruction of the injured nerve--morphological and morphometric analysis[J]. Ann Anat, 1999,181:275-281.
    [160] Arai T, Kanje M, Lundborg G, et al. Axonal outgrowth in muscle grafts made acellular by chemical extraction[J]. Restor Neurol Neurosci, 2000,17:165-174.
    [161] H.J. Jin JP, V. Karageorgiou , U.J. Kim , R. Valluzzi , P. Cebe , D. L. Kaplan Water-stable silk films with reducedβ-sheet content[J]. Advanced functional materials 2005,15:1241-1247.
    [162] Moore MJ, Friedman JA, Lewellyn EB, et al. Multiple-channel scaffolds to promote spinal cord axon regeneration[J]. Biomaterials, 2006,27:419-429.
    [163] Liu XL, Arai T, Sondell M, et al. Use of chemically extracted muscle grafts to repair extended nerve defects in rats[J]. Scand J Plast Reconstr Surg Hand Surg, 2001,35:337-345.
    [164] Yezierski RP, Liu S, Ruenes GL, et al. Excitotoxic spinal cord injury: behavioral and morphological characteristics of a central pain model [ J ] . Pain, 1998,75:141-155.
    [165] Talac R, Friedman JA, Moore MJ, et al. Animal models of spinal cord injury for evaluation of tissue engineering treatment strategies [ J ] . Biomaterials, 2004,25:1505-1510.
    [166] Sondell M, Lundborg G, Kanje M. Regeneration of the rat sciatic nerve into allografts made acellular through chemical extraction [ J ] . Brain Res, 1998,795:44-54.
    [167] Chew SY, Mi R, Hoke A, et al. Aligned Protein-Polymer Composite Fibers Enhance Nerve Regeneration: A Potential Tissue-Engineering Platform[J]. Adv Funct Mater, 2007,17:1288-1296.
    [168] Bareyre FM, Schwab ME. Inflammation, degeneration and regeneration in the injured spinal cord: insights from DNA microarrays[J]. Trends Neurosci, 2003,26:555-563.
    [169] Benowitz LI, Yin Y. Combinatorial treatments for promoting axon regeneration in the CNS: strategies for overcoming inhibitory signals and activating neurons' intrinsic growth state[J]. Dev Neurobiol, 2007,67:1148-1165.
    [170] Jiang S, Ballerini P, Buccella S, et al. Remyelination after chronic spinal cord injury is associated with proliferation of endogenous adult progenitor cells after systemic administration of guanosine[J]. Purinergic Signal, 2008,4:61-71.
    [171] Colton CK. Implantable biohybrid artificial organs[J]. Cell Transplant, 1995,4:415-436.
    [172] Basso DM, Beattie MS, Bresnahan JC, et al. MASCIS evaluation of open field locomotor scores: effects of experience and teamwork on reliability. Multicenter Animal Spinal Cord Injury Study[J]. J Neurotrauma, 1996,13:343-359.
    [173] Schmidt CE, Leach JB. Neural tissue engineering: strategies for repair and regeneration[J]. Annu Rev Biomed Eng, 2003,5:293-347.
    [174] Geller HM, Fawcett JW. Building a bridge: engineering spinal cord repair[J]. Exp Neurol, 2002,174:125-136.
    [175] Jones LL, Oudega M, Bunge MB, et al. Neurotrophic factors, cellular bridges and gene therapy for spinal cord injury[J]. J Physiol, 2001,533:83-89.
    [176] Barres BA, Raff MC, Gaese F, et al. A crucial role for neurotrophin-3 in oligodendrocyte development[J]. Nature, 1994,367:371-375.
    [177] Cohen RI, Marmur R, Norton WT, et al. Nerve growth factor and neurotrophin-3 differentially regulate the proliferation and survival of developing rat brain oligodendrocytes[J]. J Neurosci, 1996,16:6433-6442.
    [178] McTigue DM, Horner PJ, Stokes BT, et al. Neurotrophin-3 and brain-derived neurotrophic factor induce oligodendrocyte proliferation and myelination of regenerating axons in the contused adult rat spinal cord[J]. J Neurosci, 1998,18:5354-5365.
    [179]王大伟,孟晓婷,娄小倩, et al.大鼠羊膜上皮细胞植入损伤脊髓后的存活与迁移[J].中国组织工程研究与临床康复, 2007,11:2994-2996,3000.