氯化锂联合脐血干细胞及脊髓去细胞支架移植修复大鼠脊髓损伤的实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
脊髓损伤(spinal cord injuries, SCI)是现在交通、工矿事故、运动意外以及战伤中常见的损伤,伤者多为青壮年,损伤造成的截瘫不仅严重影响患者的身心健康,而且给家庭和社会造成巨大的经济、人力和精神负担。长期以来脊髓损伤的修复一直是困扰人类健康的难题,也是世界各国科学家们致力于研究的焦点,但目前缺少切实可行的有效治疗方法。脊髓损伤的治疗一直是神经科学研究的难点和热点,传统观点认为中枢神经系统损伤后,由于神经元自身再生能力差和外在微环境的抑制,损伤神经是不可再生的。1981年神经学家在基础研究中证实中枢神经损伤后的神经结构修复是可能的,掀起了各国学者对脊髓修复研究的又一次热潮。SCI后轴突再生障碍的原因相当复杂,其主要原因有:(1)对脊髓的直接损伤及继发炎症使脊髓内功能神经元大量坏死或凋亡且神经元再生困难;(2)脊髓损伤后暴露的髓鞘相关抑制分子、轴突生长抑制性蛋白的大量分泌和释放及继发形成的胶质瘢痕阻碍了轴突的生长和正确连接;(3)损伤造成局部细胞凋亡,致使细胞分泌的神经营养因子减少,破坏了神经元修复和支持轴突再生的有利微环境。目前SCI的修复研究主要集中在以下几个方面:(1)药物治疗:如传统的大剂量皮质类固醇激素(甲基强的松龙)的冲击治疗,可以有效的减轻脊髓的继发性炎症损伤,保护并防止残存的损伤神经元进一步凋亡。目前研究的氯化锂(LiCl)、免疫抑制剂FK506等除了可以保护损伤神经元外,还可以有效的促进损伤轴突再生。(2)细胞移植:细胞移植是目前研究的热点,其原理是利用某些种子细胞可以分泌多种神经营养、黏附和趋化因子,营养并保护损伤神经元、诱导轴突再生及再髓鞘化来修复SCI。既往的研究已为SCI修复提供了较成熟的种子细胞,如:胚胎干细胞(ESCs)、雪旺氏细胞(SCs)、嗅鞘细胞(OECs)、神经干细胞(NSCs)、骨髓间充质干细胞(BMSCs)、脐血干细胞(UCBCs)等,大量研究证实细胞移植均能够在一定程度上促进脊髓神经功能的恢复。(3)组织移植:随着组织工程学的飞速发展,一种更先进的SCI修复理念逐步形成,即修复SCI必须包括组织水平上的重建,整合SCI治疗研究的各种有效策略,在诱导轴突定向再生的同时,减少局部胶质瘢痕形成。目前,应用于脊髓的组织工程支架主要有两种,一种为人工合成可降解高分子生物材料,如:聚乳酸、聚乙醇酸及其共聚物等,另一种为自体或异体组织移植,如异体胚胎脊髓组织、自体肌纤维支架、游离周围神经或带血管蒂的游离周围神经组织移植修复脊髓损伤,也取得了一定的修复效果。(4)改善脊髓损伤局部微环境:SCI后轴突再生困难的另一关键因素就是适宜轴突再生的微环境遭到破坏,各种不利于轴突再生的髓磷脂源性蛋白(myelin, MAG, Nogo-A, Omgp)轴突再生抑制分子大量分泌,而各种神经营养因子(BDNF、GDNF、NGF、NT3等)的缺乏及胶质瘢痕形成阻碍了轴突再生。(5)联合治疗:采取联合不同作用机制的治疗方法,更好的发挥协同作用来促进SCI后神经修复。本研究拟用氯化锂联合脐血干细胞及脊髓去细胞支架移植修复大鼠脊髓全横断损伤,观察该方法对SCI后神经再生和脊髓修复的影响,并探讨其促进脊髓修复的作用机制。
     目的
     1.探讨脐血间充质干细胞(MSCs)体外分离、纯化、扩增的方法与条件,以及向神经细胞定向诱导分化的可行性,为脐血干细胞移植治疗脊髓损伤提供理论依据。
     2.探讨氯化锂体外诱导人脐血间充质干细胞(MSCs)向神经细胞分化的可行性。
     3.制备脊髓去细胞支架,观测脊髓内基质骨架的结构特点。
     4.探索氯化锂联合脐血干细胞及脊髓去细胞支架移植对大鼠脊髓全横断损伤的效果及其机制。
     方法
     1.无菌条件下收集正常足月儿的脐带血,肝素抗凝,密度梯度离心法分离人脐血单个核细胞,贴壁法纯化,用含15%FBS的低糖DMEM培养基进行扩增培养,流式细胞仪检测表面抗原。20ng/ml bFGF诱导MSCs向神经细胞分化,免疫组化鉴定神经元特异性标志蛋白MAP-2。
     2.取3代的脐血MSCs分3组进行体外诱导,A组:用含15%FBS和20ng/mlbFGF的DMEM完全培养基预诱导24 h,3mol/LLicl的DMEM培养基继续诱导6d;B组:含3mol/L Licl的DMEM培养基诱导7d;C组:含15% FBS的DMEM培养基正常培养7d。光镜下观察细胞形态,用免疫组化技术检测细胞NSE、MAP-2及GFAP的表达。
     3.取健康成年SD大鼠脊髓数段,每段长约2cm,手术显微镜去除脊髓表面的脂肪组织和硬脊膜,分为A、B、C三个组,每组取1个标本作为正常对照,其余段均行化学萃取。A、B、C三个组的萃取振荡频率分别为80r/min、120r/min和160r/min。将标本采用Triton X-100和脱氧胆酸钠进行萃取,蒸馏水漂洗后行切片HE染色和扫描电镜观察。萃取后的脊髓置于4℃无菌PBS溶液(0.01mol/L,pH7.4)中保存备用。
     4.首先经大鼠胸T9节段制备脊髓全横断损伤模型。120只成年雌性SD大鼠随机分为6组,每组20只:①A组为对照组:仅行T9平面脊髓全横断;②B组为Licl组:脊髓全横断+Licl(腹腔注射85mg/kg,1/日);③C组为细胞移植组:脊髓全横断+UCB-SCs移植;④D组细胞支架联合移植组:脊髓全横断+支架复合hUCB-SCs移植;⑤E组为Licl+细胞移植组:脊髓全横断+hUCB-SCs移植+Licl;⑥F组为Licl+细胞支架联合移植组:脊髓全横断+支架复合hUCB-SCs移植+Licl。于术后1d、3d及每周的最后一天,应用BBB评分评价大鼠脊髓功能的恢复情况;8周后取材,通过HE染色及Brdu细胞核标记观察移植细胞的存活、迁移;通过免疫组化染色的方法,观察hUCB-SCs向神经细胞分化及表达神经纤维的情况;通过荧光金逆行追踪,观察脊髓神经纤维的再生与分布;综合全面评价治疗措施修复脊髓损伤的效果。
     结果
     1.采用密度梯度离心法成功从脐血中分离出脐血间充质干细胞,流式细胞仪检测显示细胞不表达或弱表达CD14、CD34和CD45等造血干细胞和内皮细胞标志,但显著表达神经细胞表面标志CD90和间充质干细胞表面标志CD29、CD105。经20ng/ml bFGF诱导14天后长梭形的MSCs胞体收缩,呈圆形、不规则多边形,细胞出现类似神经元形态。兔疫组化染色MAP-2呈弱阳性表达。
     2.A组与B组诱导3d后细胞即出现形态学上的改变,细胞变成不规则形,立体感增强,从胞体伸出突起。免疫组织化学和免疫荧光方法鉴定显示,诱导后的细胞能表达神经元特异性标志NSE和MAP-2,阳性表达率A组(分别为73.6±7.8%,75.5±8.5%)高于B组(分别为31.0±4.3%,33.5±5.0%)(P<0.05),而星形胶质细胞特异性标志GFAP阳性细胞较少,A、B、C三组阳性表达率分别为4.7±3.3%、5.1±4.6%、8.5±3.2%。
     3.采用化学萃取方法可以成功制备大鼠脊髓去细胞支架,其扫描电镜结果显示,采用120r/min的震荡频率萃取的脊髓去细胞支架内基质纤维结构保存较好,基质纤维走形与脊髓纵轴一致,呈波浪状平行排列,彼此之间有短横向的基质纤维相互连接成三维镂空的网状结构。
     4.术后8周C、D、E、F组可观察至Brdu标记的脐血干细胞在体内存活并在脊髓内迁移,其中D、F组细胞存活数量多于C、E组(P<0.05)。FITC荧光标记NF-200阳性神经纤维的表达,在A组和B组未见表达;D、F组阳性表达数高于C、E组(P<0.05),可见少量连续性神经纤维通过损伤区。荧光金逆行脊髓追踪显示E组和F组在脊髓损伤区头侧中有少量被荧光金标记的神经锥体细胞,而C组和D组偶见被荧光金标记的神经锥体细胞,对照组和单纯氯化锂组未见被荧光金标记的神经锥体细胞。后肢功能运动BBB评分除对照组和单纯氯化锂组间无统计学差异外,E组和F组的BBB评分较其他组提高(P<0.05),其中,F组好于E组,D组好于C组。
     结论
     1.脐血中可以分离并扩增MSCs;脐血MSCs经bFGF体外诱导可以向神经元样细胞分化,并部分表达神经元特异性标志MAP-2。
     2.Licl体外可诱导人脐血MSCs分化为神经元样细胞,结合生长因子bFGF预诱导可大大提高其诱导效果。
     3.采用化学萃取方法可以成功制备大鼠脊髓去细胞支架;脊髓去细胞支架保存有天然的脊髓内基质纤维骨架,可有效的引导神经细胞和神经纤维定向生长和迁移。
     4.氯化锂能促进人脐血间充质干细胞在损伤区的存活并向神经细胞分化,脊髓去细胞支架内天然的脊髓基质骨架能为脊髓两侧残端基质提供良好对接,阻止外源性瘢痕长入,引导神经细胞和神经轴突定向生长和迁移,氯化锂联合脐血干细胞与脊髓去细胞支架移植能够促进细胞移植修复大鼠脊髓损伤的效果。
Spinal cord injury(SCI) is a common injury in transport, mining accidents, sports injuries and warfare nowadays. Most of the injured are young people. The paraplegia caused by SCI not only seriously affects the physical and mental health of patients, but also put up to families and the community tremendous economic, human and spiritual burden. For a long time the repair of spinal cord injury has been plaguing human health problems, and the world scientists are working on the focus, but still lack of practical and effective therapy methods. Curing spinal cord injury is always a difficult and hot point in neuroscience research. It's believed that normal mature mammalian central nervons system couldn't regenerate after injured, because of poor regeneration capability of neuron and the inhibitor form glial microenvironment. Since 1981 the neuroscientist confirmed that the repair of the central nervous system structure after injury was possible, the study on repair of SCI has been becoming hot spot in many academic institute. The reasons that interfering axon regeneration after SCI were extremely complicated. First, primary and secondary injury on spinal cord caused a great of spinal cord functional neurons apoptosis and necrosis, which making the regeneration of axons difficulty. Second, a great quantity secretion and release of myelin-associated inhibitor and axon growth inhibition protein after SCI. The secondary glial scar impeded the axons growth and properly connected. Third, the decreased secretion of many neurotrophic factors resulting from partial cells apoptosis destroyed the micro-environment benefiting for neurons repair and axon regeneration. The measures of repairing SCI currently are mainly the following aspects:drug treatment, cell transplantation, tissue transplantation, improve the local micro-environment and union treatment. The traditional high-dose corticosteroids(methylprednisolone) impacting treatment could effectively reduce the secondary inflammation after SCI, and protect the residual damaging neurons and prevent the further apoptosis. At present, the studies claimed that lithium chloride(Licl)and the immunosuppressant FK506, and so on, could protect neurons from injury and could effectively promote axon regeneration. Cell transplantation is currently a hot research, the principle is to use some seed cells secreting a variety of neurotrophic, adhesion agents, nutrite and protect the neuron after injury, and then induced axon regeneration and re-myelinization to repair SCI. Previous studies have provided a more mature seeds cells for SCI, such as: embryonic stem cells(ESCs), schwann cells(SCs), olfactory ensheathing cells (OECs), neural stem cells NSCs), bone marrow mesenchymal stem cells(BMSCs), the umbilical cord blood stem cells(UCBCs). A large number of studies have confirmed that cell transplantation facilitates the restoration of spinal cord function. With the tissue engineering rapid development, a more advanced concept of SCI repairment appeared gradually that repair of SCI must include the organization levels reconstruction. Integrating effective treatments of SCI repair strategy, we could reduce glial scar formation at the same time induce the directional axon regeneration. At present, there were two engineering scaffolds applicating in spinal cord tissue, one was biodegradable polymer synthetic biological material, such as polylactic acid, and polyglycolic acid copolymer, another was autologous or allogeneic tissue transplantation such as allogeneic embryonic spinal cord tissue, autologous muscle fiber frame, free of peripheral nerve(FPN), or the free vascularized peripheral nerve(VPN) tissue transplantation to repair spinal cord injury, and achieved a certain degree of effect. Another key factor for axon regenerate obstacle after SCI is the appropriate axonal regeneration micro-environment has been damaged, and myelin-derived protein(myelin,MAG, Nogo-A, Omgp) which inhibited the axon regeneration were greatly secreted, and lacking of various neurotrophic factor (BDNF and GDNF, NGF, NT3)and the glial scar formation hindered axon regeneration. To this end, a new therapy strategy Combined with several methods in different mechanism was used, which may have a better synergy to promote nerve repair after SCI. Accordingly,this study was designed to apply lithium chloride combined with transplantation of human umbilical cord blood mesenchymal stem cells and acellular spinal cord scaffold to repair SCI. From this study, the nerve regeneration and function recovery of SCI animal will be investigated, at the same time, the possible mechanism of recovery in SCI will be discussed.
     Obsjective:
     1. To investigate the method and conditions of isolation, proliferation of multipotent mesenchymal stem cells(MSCs)from human umbilical cord blood in vitro, and he possibility of inducing human umbilical cord blood mesenchymal stem cells (MSCs) to differentiate into neuron-like cells.
     2. To investigate the effects and possible mechanisms of treating complete transected spinal cord in rats through lithium chloride combined with transplantation of human umbilical cord blood mesenchymal stem cells and acellular spinal cord scaffold
     3. To explore a method for fabricating the acellular spinal cord scaffold and to observe the construction features of the scaffold.
     4. To investigate the possibility of inducing human umbilical cord blood mesenchymal stem cells (MSCs) to differentiate into neuron-like cells by lithium chloride (LiCl) in vitro.
     Methods:
     1. Human umbilical cord blood was collected from mature neonates. All samples were obtained sterilely with 20 U/ml heparin. The cord mononuclear cells were isolated with lymphocyte separation medium (density 1.077g/ml), then purified by wall sticking screening and expanded with slight sugar DMEM containing 15%FBS. Immunophenotypes of the cells surface were analyzed by flow eytometry. Expanded MSCs were induced to differentiate into neuron-like cells in medium added with basic fibroblast growth factor(bFGF), and immonohistochemical staining was used to identify the specific protein of neuron:microtubule associated protein-2(MAP-2)。
     2. The third passage of the expanded MSCs were pre-inducted with DMEM containing 15%FBS and 20ng/ml bFGF for 24 hours, then induced with DMEM without serum but 3mol/L Licl for 6 days in group A. The MSCs were induced with DMEM containing 3mol/L Licl for 7 days in group B. The MSCs were normally cultured with DMEM containing 15%FBS in group C. The morphological changes of the cells were observed under phase contrast microscope. The neuron specific markers containing neuron specific enolase(NSE), microtubule associated protein-2(MAP2) and glial fibrillary acid protein(GFAP) were evaluated by indirect immunocyto-chemistry staining.
     3. Several Segments of spinal cord were obtained from Adult female Sprague-Dawley rats. Under operative microscopy, the fat and a part of dural matter were cutted before the extraction procedure. Segments of spinal cord obtained from rats were divided randomly into three groups: group A(frequency of vibration=80r/min), group B(frequency of vibration=120r/min) and group C (frequency of vibration=160r/min). The spinal cord Was delt with solution of Triton X-100 and with solution of sodium deoxycholate at room temperature. Then washed with distilled-water, delt with HE staining and observed under light microscope. Scanning electron microscope was used to observe the ultramicrostructure. The fabricated acellular spinal cord scaffolds were stored in 0.01mol/L PBS(PH7.4).
     4. At first, to establish the entire transected spinal cord injury model at T9 level in rats. Then 120 Sprague-Dawley (SD) female rats were divided randomly into six groups,20 in each group. Group A(spinal cord entire transaction), group B (spinal cord entire transaction+intraperitoneal injection of 85mg/kg lithium chloride every day), group C (spinal cord entire transaction+hUCB-SCs transplantation), group D (spinal cord entire transaction+hUCB-SCs transplantation+intraperitoneal injection lithium chloride), group E (spinal cord entire transaction+acellular spinal cord scaffold and hUCB-SCs transplantation), group F (spinal cord entire transaction+ acellular spinal cord scaffold and hUCB-SCs transplantation+intraperitoneal injection lithium chloride). At 1 day,3day and the end day of every week post-operation, a behavioral testing was performed weekly upon each hindlimb of all animals according to the BBB scoring system. At the 8th week, all animals were sacrificed and the spinal cords were taken out for morphological observation. Tissues in SCI sites were observed with H&E staining and Brdu nuclear labeling to identify the survival and migration of SCs. With immunocyto-chemistry staining to identify the differentiation of neuron-like cells and expression of the neurofibras. With fluorescent-gold(FG) spinal cord retrograde tracing to observe the regeneration and distribution of spinal nerve fiber. The treatment effects of spinal cord injury were identified comprehensively.
     Results:
     1. The hUCB-SCs could be isolated successfully from the UCB with density gradient centrifugation. Flow cytometry analysis showed that the cells were positive for CD90 (neural cell antigen)and CD29, CD105 (MSC-specific surface markers), while negative for CD34 (hematopoietic stem cell antigen)and CD45 (leukocyte common antigen). MSCs differentiated into neuron-like cells induced for 14 days by 20ng/ml bFGF and the differentiated cells faintly expressed MAP-2.
     2. After inducted for 3 days, morphological changes were observed obviously in group A and B.6 days later, the differentiated cells showed typical neuronal morphology. The expression of NSE and MAP2 were positive for the majority cells in group A and B, but that of group A [(73.6±7.8)%, (75.5±8.5)%]were obviously higher than group B [(31.0±4.3)%, (3.5±5.0)%]. few expressed GFAP in both groups.
     3. Acellular spinal cord scaffold can be fabricated by chemical extraction. The three-dimensional(3D)structure of the acellular spinal cord scaffold are kept intact, in which there are bundles of extracellular matrix (ECM) fiber which aligned lengthways were interlaced with transversal matrix fibers.
     4. It could be observed that Brdu marked hUCB-SCs survivaed and migrated in the spinal cord postoperative 8th week in groupC, D, E and F. The survivaed hUCB-SCs in group D and F were extremely more than that in group C and E. Through FITC fluorescence labeling NF-200 positive nerve fibers, No NF-200 positive nerve fibers could be observed in group Aand B; NF-200 positive nerve fibers in group D and F were extremely more than that in group C and E. Some continuited nerve fibers through injury district could be observed in group D and F. Fluorogold(FG)retrograde tracing show that a small amount of pyramidal cells were labeled by FG in group E and F. The nerve pyramidalcells marked by FG occasionally were observed in group C and D, the remaining two groups were not apperence. There were significant differences of BBB Score of hindlimb functional movement among all groups except group Aand B. The BBB Score of hindlimb functional movement were better in group F than that in group E, better in group D than that in group C.
     Conclusions:
     1. The results suggest that MSCs can be obtained from HUCB. MSCs from HUCB can be induced to differentiate into neuron-like cells and faintly express MAP-2 in vitro.
     2. Licl could induce the human umbilical cord blood MSCs to differentiate into neuron-like cells in vitro. Licl combined with bFGF could improve the induced effects.
     3. Acellular spinal cord scaffold can be fabricated by chemical extraction. The native 3D structure of the ECM in the scaffold maybe provide the structural foundation for inducing effectively the neurons and axons to directionally grow and migrate.
     4. Licl could improve survival and differentiation into neural cells of the human umbilical cord blood in injury district. Acellular spinal cord scaffold could bridge the both stumps of the injured spinal cord by means of "alete butt joint", stop the in-migrating of the peripheral tissues around the spinal cord, guide the directional growth and migration of the neural cells and axons. Licl combined with transplantation of human umbilical cord blood mesenchymal stem cells and acellular spinal cord scaffold could improve the functional recovery of the hindlimbs in the complete transected spinal cord rats.
引文
[1]Ohgushi H, Caplan AI. Stem cell technology and bioceramics:from cell to gene engineering[J]. J Biomed Mater Res.1999,48:913-927
    [2]Dale Woodbury, Emily J. Schwarz, Darwin J. Prockop, et al. Adult Rat and Human Bone Marrow Stromal Cells Differentiate Into Neurons[J]. Journal of Neuroscience Research,2000,61:364-370
    [3]J. Sanchez-Ramos, S. Song, F. Cardozo-Pelaez, et al. Adult Bone Marrow Stromal Cells Differentiate into Neural Cells in intro[J]. Experimental Neurology, 2000,164:247-256
    [4]D'Ippolito G, Schiller PC, Ricordi C, et al. Age-related osteogenic potential of mesenchymal stromal stem cells from human vertebral bone marrow[J]. J Bone Miner Res,1999,14:1115-1122
    [5]Erices A, Conget P, Minguell J. Mesenchymal progenitor cells in human umbilical cord blood[J]. Br J Haemato 1,2000,109:235-242
    [6]Mareschi K, Biasin E, Piacibello W,et al. Isolation of human mesenchymal stem cells:bone marrow versus umbilical cord blood[J]. Haematologica,2001,86: 1099-1100
    [7]Goodwin H, Bicknese A, Chien S, et al. Multilineage differentiation activity by cells isolated from umbilical cord blood: expression of bone, fat, and neural markers[J]. Biol Blood Marrow Transplant,2001,7:581-588
    [8]Friedenstein AJ, Piatetzyky-Shapiro I, Petrovka KV Osteogenesis in transplants of bone marrow cells [J]. J Embryol Exp Morph,1966,16:38190.
    [9]Kopen GC, Prockop DJ, Marrow stromal cells migrate throughout forebrain and cerebellum, and they differentiate into astrocytes after injection into neonatal mouse brains. Proc Natl Acad Sci USA,1999,96:10711-10716.
    [10]Zhao LR,Duan WM, Reyes M, et al. Human bone marrow stem cells exhibit neurological deficits after grafting into the chemic brian of rats[J]. Exp Neurol, 2002,174:11-20
    [11]Hofstetter CP, Schwarz EJ, Hess D, et al. Marrow stromal cells form Guiding strands in the injured spinal cord and promote recovery. Proc Natl Acad Sci USA, 2002.99:2199-2204.
    [12]Satake K, Lou J, Lenke LG. Migration of mesenchymal stem cells through cerebrospinal fluid into injured spinal cord tissue. Spine 2004,29:1971-1979.
    [13]Daniel P. Ankeny, Dana M. Mc Tigue, et al. Bone marrow transplants provide tissue protection and directional guidance for axons after contusive spinal cord injury in rats. Experimental Neurology.2004,190(1):1731.
    [14]Bianco P, Robey PG. Marrow stromal stem cells [J]. J Clininvest,2000,15 (12):1663~1668.
    [15]Pittenger MF,Mackay AM,Beck SC,et al.Multilineage potential of adult human mesenchymal stem cell[J]. Science,1999,284:143~147.
    [16]Petersen BE, Bowen WC. Bone marrow as a source of hepatic oval cells [J]. Science,1999,284:1168-1170.
    [17]Makino S, Fukuda K, Migoshi S, et al.Cardiomyocytes can be generated from marrow stromal cells in vitro[J]. J Clin Invest,1999,103 (5):697-705.
    [18]Pereira RF, O'Hara MD, Halford KW, et al.Cultured adherent cells from marrow can serve as long lasting precursor cells for bone and lung in irradiated mice[J].Proc Natl Acad Sci USA,1995,92:4857-4861.
    [19]O'Donoghue K, Choolani M, Chan J, et al. Identification of fetal mesenchymal stem cells in maternal blood: Implications for non-invasive prenatal diagnosis[J]. Mol Hum Reprod,2003,9:497-502.
    [20]Zvaifler NJ, Marinova-Mutafchieva L, A dams G, et al. Mesenchymal precursor cells in the blood of normal individuals[J]. Arthritis Res,2000,2:477-488.
    [21]Oscar K. Lee, Tom K. Kuo, Wei-Ming Chen, et al. Isolation of multipotent mesenchymal stem cells from umbilical cord blood[J]. BLOOD, 2004,103:1669-1675.
    [22]J. W. Kim, S. Y. Kim, S. Y. Park, et al. Mesenchymal progenitor cells in the human umbilical cord, [J]. Ann Hematol,2004,83:733-738.
    [23]Myoung Woo Lee, a Jeongeun Choi, a Mal Sook Yang, et al. Mesenchymal stem cells from cryopreserved human umbilical cord blood [J].Biochemical and Biophysical Research Communications,2004,320:273-278.
    [24]侯玲玲曹华裴雪涛等。人脐血间充质干细胞体外扩增和向神经元样细胞定向诱导分化的研究[J].中华血液学杂志,2002,23(8):415-419.
    [25]Tavassoli, M. Embryonic and fetal hemopoiesis:an overview[J]. Blood Cells, 1991,1:269-281.
    [26]Peault, B. Hematopoietic stem cell emergence in embryonic life:developmental hematology revisited[J]. Journal of Hematotherapy,1996,5:369-378.
    [27]Woodbury D, Schwarz EJ, Prockop DJ, et al. Adult rat and human bone marrow stromal cells differentiate into neurons[J]. J Neurosci Res,2000,61:364-370.
    [28]J. Sanchez-Ramos, S. Song, F. Cardozo-Pelaez, et al. Adult Bone Marrow Stromal Cells Differentiate into Neural Cells in Vitro [J]. Experimental Neurology,2000,164:247-56.
    [29]Deng W, Obrocka M, Fischer I, et al. In vitro differentiation of human marrow stromal cells into early progenitors of neural cells by conditions that increase intracellular cyclic AMP Biochem Biophys Res Commun,2001,282:148-52.
    [30]Burkhalter J, Fiumelli H, Allaman I, et al. Brain-derived neurotrophic factor stimulates energy metabolism in developing cortical neurons [J]. J Neurosci, 2003,23:8212-20.
    [1]Romanov YA,Svintsitskaya VA,Smimov VN. Searching for alternative sources of postnatal human mesenchymal stem cells:candidate MSC-like cells from umbilical cord. Stem Cells 2003,21:105-110
    [2]Lee OK,Kuo TK,Chen WM,et al.Isolation of multipotent mesenchymal stem cells from umbilical cord blood.Blood 2004,103:1669-1675
    [3]Su. Kuh, Y.E. Cho, D.H. Yoon, K.N. Kim and Y. Ha, Functional recovery after human umbilical cord blood cells transplantation with brain-derived neutrophic factor into the spinal cord injured rat, J Acta Neurochir,2005,147:985-992.
    [4]Kim JS, Chang MY, Yu IT, et al. Lithium selectively increases neuronal differentiation of hippocampal neural progenitor cells both in vitro and in vivo, J Neurochem Res,2004,89:324-336.
    [5]Su HX, Chu TH, and Wu WT, Lithium enhances proliferation and neuronal differentiation of neural progenitor cells in vitro and after transplantation into the adult rat spinal cord. J Experimental Neurology,2007,206:296-307.
    [6]Longo FM, Yang T, Xie Y, et al. Small molecule approaches promoting neurogenesis [J]. Curr Alzheimer Res.2006 Feb,3:5-10.
    [7]Rebecca Owen, Phillip R Gordon-Weeks. Inhibition of glycogen synthase kinase 3β in sensory neurons in culture alters filopodia dynamics and microtubule distribution in growth cones [J]. Mol Cell Neurosci.2003 Aug,23:626-637.
    [8]Lenox RH. Wang L. Molecular basis of lithium action:integration of lithium-responsive signaling and gene expression networks [J]..Mol Psychiatry.2003,8:135-144.
    [9]Bauer M, Alda M, Priller J, et al. Implications of the neuroprotective effects of lithium for the treatment of bipolar and neurodegenerative disorders [J]. Pharmacopsychiatry.2003,36:250-254.
    [10]Yick LW, So KF, Cheung PT, et al. Lithium chloride reinforces the regeneration-promoting effect of chondroitinase ABC on rubrospinal neurons after spinal cord injury[J]. J Neurotrauma.2004,21:932-943.
    [11]Huang X, Wu DY, Chen G, et al. Support of retinal ganglion cell survival and axon regeneration by lithium through a Bcl-2-dependent mechanism[J]. Invest ophthalmol Vis Sci.2003,44:347-354.
    [12]Alvarez G, Munoz-Montano JR, Satrustegui J, et al. Regulation of tau phosphorylation and protection against beta-amyloid-induced neurodegeneration by lithium, Possible implications for Alzheimer'S disease[J]. Bipolar Disord.2002,4:153-165.
    [13]Manji HK, Moore GJ, Chen G. Lithium up-regulates the cytoprotective protein Bcl-2 in the CNS in vivo:a role for neurotrophic and neuroprotective effects in manic depressive illness[J]. J ClinPsychiatry.2000,61:82-96.
    [14]卞清明,钱燕宁.Licl对沙土鼠前脑缺血后神经细胞凋亡及磷酸化Akt蛋白表达的影响.南京医科大学学报.2005,25:540-543.
    [15]侯永根,端礼荣,吴全义等.氯化锂对大鼠胚胎中脑神经细胞毒性作 用的研究.苏州大学学报.2003,13:323-326.
    [16]Lim JY, Park SI, Oh JH et al. Brain-derived neurotrophic factor stimulates the neural differentiation of human umbilical cord blood derived mesenchymal stem cells and survival of differentiated cells through MAPK/ERK and PI3K/Akt-dependent signaling pathways. J Neurosci Res,2008,86:2168-2178
    [17]Kamishina H, Deng J, Oji T, et al. Expression of neural markers on bone marrow-derived canine mesenchymal stem cells. Am J Vet Res.2006, 67:1921-1928.
    [18]G. Chen,G. Rajkowska,F. Du, N. Seraji-Bozorgzad and H.K. Manji, Enhancement of hippocampal neurogenesis by lithium, J Neurosci Res,2000,75: 1729-1734
    [19]Tureyen K, Vemuganti R, Bowen KK et al. EGF and FGF infusion increases post-ischemic neural progenitor cell proliferation in the adult rat brain. J Neurosurgery,2005,57:1254-1263
    [20]Jeong JA, Gang EJ, Hong SH, et al. Rap id neural differentiation of human cord blood-derived mesenchymal stem cells. J Neuroreport,2004,15:1731-1734.
    [21]袁源,杨树源,韩忠朝,等.人脐带间充质干细胞体外扩增和向神经元样细胞定向诱导分化的研究.中华神经医学杂志,2006,5:230-236.
    [22]夏桂枝,张国成,陈新民,等人脐血间充质干细胞体外终末分化为神经元样细胞.细胞与分子免疫学杂志,2008,24:741-743.
    [1]Sondell M, Lundborg G, Kanje M. Regeneration of the rat sciatic nerve into allografts made acellular through chemical extraction[J]. Brain Res,1998, 795(1-2):44-54.
    [2]Langer R, Tirrell DA. Designing materials for biology and medicine. Nature,2004,428:487-492.
    [3]Novikova LN, Kellerth JO, Novikov LN. Biopolymers and biodegradable smart implants for tissue regeneration after spinal cord injury. Curr Opin Neurol,2003,16(6):711-715.
    [4]MaW, FitzgeraldW, LiuQY, et al. CNS stemand progenitor cell differentiation into functional neuronal circuits in three-dimensional collagen gels. Exp Neurol,2004,190:276-288.
    [5]Yoshii S, Oka M, Shima M, et al. Bridging a spinal cord defect using collagen filament. Spine,2003,28(20):2346-2351.
    [6]Flemming RG, Murphy CJ, Nealey PF, et al. Effects of synthetic micro-and nano-structured surfaces on cell behavior. Biomaterials,1999,20: 573-588.
    [7]LutolfMP, Hubbell JA. Synthetic biomaterials as instructive extracellular microenvironments for morphogenesis in tissue engineering. Nature Biotechnology,2005,23:47-55.
    [8]Silva GA. Introduction to nanotechnology and its applications to medicine. Surg Neurol,2004,61:216-220.
    [9]Silva GA. Neuroscience nanotechnology:progress, opportunities and challenges. Nat Rev Neurosci.2006,7:65-74.
    [10]Yang F, Murugan R, Ramakrishna S, et al. Fabrication of nanostructured porous PLLA scaffold intended for nerve tissue engineering. Biomaterials, 2004,25(10):1891-1900.
    [11]Yang F, Murugan R, Wang S, et al. Electrospinning of nano/micro scale poly (L-lactic acid) aligned fibers and their potential in neural tissue engineering. Biomaterials,2005,26(15):2603-2610.
    [12]Yang F, Xu CY, Kotaki M, et al. Characterization of neural stem cells on electrospun poly (L-lactic acid) nanofibrous scaffold. J Biomater Sci Polym Ed,2004,15(12):1483-1497.
    [13]Kenawy el, R, et al. Electrospinning of poly (ethylene-covinyl alcohol) fibers. Biomaterials,2003,24:907-913.
    [14]Zhang S. Fabrication of novel biomaterials through molecular self-assembly. Nature Biomaterials,2003,21:1171-1178.
    [15]Estroff LA, Hamilton AD. Water gelation by small organicmolecules. Chem. Rev,2004,104:1201-1218.
    [16]Holmes TC, SuXing, Zhang S, et al. Extensive neurite outgrowth and active synapse formation on self-assembling peptide scaffolds. Proc. Natl. Acad. Sci.2000,97:6728-6733.
    [17]Pakstis LM, Bulent Ozbas, Hales KD, et al. Effect of chemistry and morphology on the biofunctionality of self-assembling diblock copolypetide hydrogels. Biomacromolecules,2004,5(2):312-318.
    [18]Silva GA, Czeisler C, Niece KL, et al. Selective differentiation of neural progenitor cells by high-epitope density nanofibers. Science,2004,303: 1352-1355.
    [19]Silva GA. Nanotechnology approaches for the regeneration and neuroprotection of the central nervous system. Surgical Neurology,2005, 63:301-306.
    [20]Niece KL, Hartgerink JD, Donners JJ, et al. Self-assembly combining two bioactive peptide-amphiphile molecules into nanofibers by electrostatic attraction. Journal of the American Chemical Society.2003,125 (24): 7146-7158.
    [21]Schneider JP, PochanDJ, Ozbas B, et al. Responsive hydrogels from the intramolecular folding and self-assembly of a designed peptide. J Am Chem Soc 2002,124:15030-15037.
    [22]Curtis ASG, Gadegaard N, Dalby MJ, et al. Cells react to nanoscale order and symmetry in their surroundings. IEEE TransNanobioscience 2004,3: 61-65.
    [23]Pochan DJ, Schneider JP, Kretsinger J, et al. Thermally reversible hydrogels via intramolecular folding and consequent self-assembly of a de novo designed peptide. J Am Chem Soc 2003,125:11802-11803.
    [24]郭树章,任先军,蒋涛,等.脱细胞脊髓天然支架的制备及形态学观察[J].中国矫形外科杂志,2007,15(3):225-228
    [25]Rovak JM, Bishop DK, Boxer IX, et al. Peripheral nerve transplantation: the role of chemical acelularization in eliminating allograft antigenicity[J]. J Reconstr Microsurg,2005,21(3):207-213.
    [26]孙明学,唐金树,王鑫,等.去细胞处理对化学去细胞异体神经免疫原性的影响[J].中华外科杂志,2006,44(4):275-278
    [27]Hudson TW, Zawko S,Deister C, et al. Optimized acellular nerve graft is immunologically tolerated and supports regeneration[J]. Tissue Eng,2004, 10 (11-12):1641-1651.
    [28]陈秉耀,侯树勋,衷鸿宾,等.兔化学去细胞神经移植修复大鼠坐骨神经缺损[J].中国矫形外科杂志,2003,11(21):1476-1478.
    [29]Kim BS, Yoo JJ, Atala A. Peripheral nerve regeneration using acellular nerve grafts[J]. J BiomedMater ResA,2004,68(2):201-209.
    [30]郭树章,任先军,蒋涛,等.大鼠脱细胞异体脊髓支架的免疫原性研究 [J].中国矫形外科杂志,2007,15(16):444-446.
    [31]Gulati A K. Immunological fate of Schwann cell populated acellular basal lamina nerve allografts[J]. Transplantation,1995,59(11):1618-1622.
    [32]Friedman JA, Windebank AJ, Moore MJ, et al. Biodegradable polymer grafts for surgical repair of the injured spinal cord[J]. Neurosurgery,2002, 51:742-751.
    [33]Won BK, Tetzlaff W. Spinal cord regeneration from gene to transplants[J]. Spine,2001,26(24Suppl):13-22.
    [34]Ribatti D, Conconi MT, Nico B, et al. Angiogenic response induced by aeellular brain scaffolds grafted onto the chick embryo chorioallantoic membrane[J]. Brain Research,2003,989(1):9-15.
    [35]Zhang L, Sirivisoot S, Balasundaram G, et al. Nanoengineering for bone tissue engineering. In:Khademhosseini A, editor. Micro-and nanoengineering of the cell microenvironment:technologies and applications[M]. Norwood: Aaech House,2008, p431-460.
    [36]Christenson EM, Anseth KS, Vall den Beucken JJJP, et al. Nanobiomaterial applications in orthopedics[J]. J Orthop Res,2007,25:11-22.
    [37]Chun AL, Moralez JG, Webster TJ, Fenniri H. Helical rosette nanotubes:a biomimetic coating for orthopedics[J]. Biomaterials,2005,26:7304-7309.
    [38]Zhang L, Ramsaywack S, Fenniri H, et al. Enhanced osteoblast adhesion on self-assembled nanostructured hydrogel scaffolds[J]. Tissue Eng,2008, 14:1353-1364.
    [39]Motta A, Fambri L, Migliaresi C. Regenerated silk fibroin films:thermal and dynamic mechanical analysis[J]. Macromolecular Chemistry and Physics,2002,203(10-11):1658-1665.
    [40]LaCroix D, Chateau A, Ginebra MP, et al. Micro-finite element models of bone tissue-engineering scaffolds[J]. Biomaterials,2006,27(30): 5326-5334.
    [41]Jones AC, Arns CH, Sheppard AP, et al. Assessment of bone ingrowth into porous biomaterials using MICRO-CT[J]. Biomaterials,2007,28(15): 2491-2504.
    [42]Oudega M, Gautier SE, Chapon P, et al. Axonal regeneration into Schwann cell grafts within resorbable poly(alpha-hydmxyacid)guidance channels in the adult rat spinal cord[J]·Biomaterials,2001,22:1125-1136.
    [43]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 USA,2002,99:3024-3029.
    [44]Blacher S, Maquet, Schils F, et al. Image analysis of the axonal ingrowth into poly(D, L-lactide)porous scaffolds in relation to the 3-D porous structure [J]. Biomaterials,2003,24:1033-1040.
    [45]Karuri NW, Liliensiek S, Teixeira AI, et al. Bilogical length scale topography enhances cell-substratum adhesion of human corneal epithelial cells[J]. Journal of Cell Science,2004,117(15):3153-3164.
    [46]Liliensiek SJ, Campbell S, Nealey PF, et al. The scale of substratum topographic features modulates proliferation of comeal epithelial cells and corneal flbroblasts [J]. Journal of Biomedical Materials Research-Part A, 2006,79(1):185-192.
    [47]Loesberg WA, te Riet J'vail Delft FCMJM, et al. The threshold at which substrate nanogroove dimensions may influence fibroblast alignment and adhesion [J]. Biomaterials,2007,28(27):3944-3951.
    [48]Karageorgiou V Kaplan D. Porosity of 3D biomaterial scaffolds and osteogenesis[J]. Biomaterials,2005,26(27):5474-5491.
    [49]Shao Z, Vollrath F. Surprising strength ofsilkworm silk[J]. Nature,2002, 418(6899):741.
    [50]Panilaitis B, Altman GH, Chen J, el al. Macrophage responses to silk [J]. Biomaterials,2003,24(18):3079-3085.
    [51]Arai T, Freddi G, Innocenti T, et al. Biodegradation of Bombyx mori silk flbroin fibers and films [J]. Journal of Applied Polymer Science,2004,91: 2383-2390.
    [52]Jin H-J, Park J, Karageorgiou V, et al. Water-stable silk films with reduced beta-sheet content[J]. Advanced Functional Materials,2005,15: 1241-1247.
    [53]Meinel L, Hofmann S, Karageorgiou V, et al. The inflammatory responses to silk films in vitro and in vivo [J]. Biomaterials,2005,26: 147-155.
    [54]Vepari C, Kaplan DL. Silk as a biomaterial[J]. Progress in Polymer Science(Oxford),2007,32(8-9):991-1007.
    [55]Lawrence BD, Cronin-Golomb M, Georgakoudi I, et ai. Bioactive silk protein biomaterial systems for optical devices[J]. Biomacromolecules,2008, 9(4):1214-1220.
    [56]Wu Y Shen Q, Hu S. Direct electrochemistry and electrocatalysis of heme-proteins in regenerated silk fibroin film[J]. Analytica ChimicaActa, 2006,558(1-2):179-186.
    [57]Hofmann S, Wong Po Foo CT, Rossetti F, et al. Silk fibroin as an organic polymer for controlled drug delivery[J]. Journal ofControlled Release,2006, 111(1-2):219-227.
    [58]Michael J.Moore, Jonathan A. Friedman, Eric B. Lewellyn, et al. Multiple-channel scaffolds to promote spinal cord axon regeneration [J]. Biomatedals,2006,27:419-429.
    [1]Zhang N, Yan HH, Wen XJ. Tissue-engineering approaches for axonal guidance. Brain res rev,2005,49:48-64.
    [2]Takezawa T. A strategy for the development of tissue engineering scaffolds that regulate cell behavior. Biomaterials,2003,24:2267-2275.
    [3]Xu XM, Guenard V Kleitman N, et al. Axonal regeneration into Schwann-cell-seeded guidance channels grafted into transected adult rat spinal cord. Comp Neurol,1995,351(1):145-160.
    [4]Guest JD, Rao A, Olson L, et al. The ability of human Schwann cell gratis to promote regeneration in the transected nude rat spinal cord. Exp Neurol,1997, 148:502-522.
    [5]N. Weidner, A. Ner, N. Salimi, M. H. Tuszynski. Spontaneous corticospinal axonal plasticity and functional recovery after adult central nervous system injury[J]. PNAS,2001,98(6):3513-3518.
    [6]Rebecca Owen, Phillip R Gordon-Weeks. Inhibition of glycogen synthase kinase 3β in sensory neurons in culture alters filopodia dynamics and microtubule distribution in growth cones [J]. Mol Cell Neurosci.2003 Aug: 23(4):626-637.
    [7]Lenox RH. Wang L. Molecular basis of lithium action:integration of lithium-responsive signaling and gene expression networks[J]. Mol Psychiatry.2003,8:135-144.
    [8]Bauer M, Alda M, Priller J. et al. Implications of the neuroprotective effects of lithium for the treatment of bipolar and neurodegenerative disorders [J]. Pharmacopsychiatry.2003,36:250-254.
    [9]Yick LW, So KF, Cheung PT, et al. Lithium chloride reinforces the regeneration-promoting effect of chondroitinase ABC on rubrospinal neurons after spinal cord injury[J]. J Neurotrauma.2004,21:932-943.
    [10]Huang X, Wu DY, Chen G, et al. Support of retinal ganglion cell survival and axon regeneration by lithium through a Bcl-2-dependent mechanism[J]. Invest ophthalmol Vis Sci.2003,44:347-354.
    [11]Alvarez G, Munoz-Montano JR, Satrustegui J, et al. Regulation of tau phosphorylation and protection against beta-amyloid-induced neurodegeneration by lithium, Possible implications for Alzheimer'S disease[J]. Bipolar Disord.2002,4:153-165.
    [12]Manji HK, Moore GJ, Chen G. Lithium up-regulates the cytoprotective protein Bcl-2 in the CNS in vivo:a role for neurotrophic and neuroprotective effects in manic depressive illness[J]. J ClinPsychiatry.2000,61:82-96.
    [13]Mitchell KE, Weiss ML, Mitchell BM, et al. Matrix Cells from Wharton'S Jelly Form Neurons and Glia[J]. Stem Cells,2003,21:50-60.
    [14]Woodbury D, Schwarz EJ, Prockop DJ, et al. Adult rat and human bone marrow stromal cells differentiate into neurons[J]. Neurosci Res,2000,61: 364-370.
    [15]Zigova T, Song S, Willing AE, et al. Human umbilical cells express neural antigens after transplantation into developing rat brain. Cell Transplant,2002, 11:265-274
    [16]Ende N, Chen R. Parkinson's disease mice and human umbilical cord blood. J-Med,2002,33:173-180
    [17]Ende N, Chen R., Ende-Harris D. Human umbilical cord blood cells ameliorate Alzheimer's disease in transgenic mice.J-Med,2001,32:241-247
    [18]Chen J, Sanberg PR, Li Y, et al. Intravenous administration of human umbilical cord blood reduces behavioral deficits after stroke in rats. Stroke, 2001,32:2682-2688
    [19]王革生,张庆俊,韩忠朝,等.人脐带间充质干细胞移植对大鼠脊髓损伤神经功能恢复的评价.中华神经外科杂志,2006,22:18-21.
    [20]蔡培强,汤逊,林月秋,等.Lentivims介导分泌神经营养因子.3的基因工程神经干细胞移植治疗脊髓损伤的实验研究.中华外科杂志,2005,43(16):1091-1093.
    [21]Friedman JA, Windebank AJ, Moore MJ, et al. Biodegradable polymer grafts for surgical repair of the injured spinal cord[J]. Neurosurgery,2002,51: 742-751.
    [22]Sondell M, Lundborg G,Kanje M. Regeneration of the rat sciatic nerve into allografts made acellular through chemical extraction[J]. Brain Res,1998, 795(1-2):44-54.
    [23]Won BK, Tetzlaff W. Spinal cord regeneration from gene to tran splants[J]. Spine,2001,26:13-22.