表皮生长因子受体抑制剂对大鼠脊髓损伤后轴突再生微环境的作用机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
第一部分表皮生长因子受体抑制剂AG1478对大鼠脊髓损伤后GAP-43的表达及运动功能缺损的影响
     目的:脊髓损伤后胶质瘢痕的形成及髓鞘相关抑制分子的产生是导致轴突不能有效再生的重要原因。而表皮生长因子受体的活化可导致静息星形胶质细胞反应性增生并形成胶质瘢痕,而且表皮生长因子受体抑制剂AG1478通过减轻髓鞘相关抑制分子及硫酸软骨素蛋白聚糖对轴突再生的抑制作用而促进了视神经轴突的再生,因此表皮生长因子受体抑制剂AG1478可能对脊髓损伤后轴突的再生有促进作用而改善脊髓损伤所导致的神经功能缺损。在本实验中我们通过在损伤脊髓局部给予表皮生长因子受体抑制剂AG1478以观察AG1478能否上调相关生长蛋白-43(GAP-43)的表达及其是否改善大鼠脊髓损伤后神经功能缺损症状。
     方法:应用重物坠落打击法建立大鼠脊髓损伤模型,假手术组仅切除T10椎板。48只健康雌性成年SD大鼠随机分为假手术组、损伤对照组和AG1478治疗组。AG1478治疗组大鼠在脊髓损伤后立即在局部给予含有0.2 ml2mM的AG1478的明胶海绵外敷治疗,而损伤对照组和假手术均在术后立即给予含溶剂(二甲基亚砜)的明胶海绵外敷作对照治疗。应用免疫荧光技术和western blot检测各组大鼠脊髓损伤后28天损伤区域生长相关蛋白-43(GAP-43)的表达情况。应用BBB评分法对大鼠脊髓损伤后第1d及以后每周(共8周)后肢运动功能进行评分。在损伤前及损伤后每周(共8周)测量各组大鼠体重。
     结果:免疫荧光检测显示在损伤后28天AG1478组GAP-43阳性细胞计数表达较对照组明显增多(P<0.01)。Western blot印迹分析也显示AG1478组GAP-43表达较对照组明显上调(P<0.01)。损伤对照组与AG1478治疗组大鼠在伤后第1天后肢均几乎完全瘫痪,表明两组大鼠损伤程度一致。经重复测量数据方差分析检验后显示AG1478组BBB评分明显高于对照组(P<0.01)。经重复测量数据方差检验分析显示AG1478组大鼠体重明显重于对照组大鼠体重P<0.05)。
     结论:脊髓损伤后立即在损伤局部给予表皮生长因子受体抑制剂AG1478干预,可以改善大鼠脊髓损伤后的神经功能缺损并明显上调与轴突生长相关的GAP-43。表皮生长因子受体抑制剂AG1478对脊髓损伤有确切的治疗作用,这提示其可能是中枢神经系统损伤的有效保护剂。
     第二部分表皮生长因子受体抑制剂AG1478对大鼠脊髓损伤后反应性星形胶质活化增生的影响
     目的:星形胶质增生被认为在脊髓损伤的病理生理改变中起着极其重要的作用,而在中枢神经系统损伤的病理生理改变中星形胶质细胞的增生与表皮生长因子受体的活化密切相关。因此我们试图应用表皮生长因子受体抑制剂AG1478对脊髓损伤进行干预以观察表皮生长因子受体抑制剂AG1478对脊髓损伤后星形胶质细胞上磷酸化表皮生长因子受体(pEGFR)的表达和反应性胶质增生及轴突生长抑制因子硫酸软骨素蛋白聚糖(CSPGs)产生的影响
     方法:应用重物坠落打击法建立大鼠脊髓损伤模型,假手术组仅切除T10椎板。45只健康雌性成年SD大鼠随机分为假手术组、损伤对照组和AG1478治疗组。AG1478治疗组大鼠在脊髓损伤后立即在局部给予含有0.2 ml2mM的AG1478的明胶海绵外敷治疗,而损伤对照组和假手术均在术后立即给予含溶剂(二甲基亚砜)的明胶海绵外敷作对照治疗。分别于损伤后14d、28d处死各组实验大鼠。应用免疫荧光技术和western blot技术检测GFAP在损伤后14天、28天的表达水平变化情况;采用免疫荧光双标技术检测脊髓损伤后14天各组大鼠星形胶质细胞表达磷酸化表皮生长因子受体(pEGFR)的情况并检测伤后28天时星形胶质细胞表达硫酸软骨素蛋白聚糖(CSPGs)的情况。
     结果:假手术组GFAP阳性细胞稀少,形态纤细;而损伤对照组在伤后14天GFAP表达明显上调,星形胶质细胞呈突触增多变大的活化状态,伤后28天典型胶质瘢痕出现;在AG1478治疗组GFAP的表达明显受到抑制,而伤后28天无明显胶质瘢痕形成。GFAP Western印迹分析也显示在损伤后14天和28天,AG1478组GFAP的表达均明显低于对照组假手术组(P<0.01)。假手术组pEGFR和GFAP表达均很弱,无荧光染色双标阳性区域,表明正常脊髓组织的星形胶质细胞不表达pEGFRo对照组pEGFR和GFAP均有强表达,同时损伤组脊髓组织病灶周围存在明显空洞和疤痕改变,且在空洞周围疤痕存在pEGFR和GFAP双标阳性区域,表明脊髓损伤后星形胶质细胞表达pEGFR,然而AG1478组pEGFR和GFAP的表达均较弱,其荧光强度较对照组明显较弱(P<0.01),且无pEGFR和GFAP双标阳性区域,表明AG1478对pEGFR和GFAP的表达均有明显抑制作用。假手术组脊髓组织CSPGs和GFAP表达均较弱;而对照组CSPGs和GFAP均有较强表达,且损伤中心CSPGs表达很强,在胶质瘢痕区域CSPGs和GFAP存在明显双标阳性区域,表明活化星形胶质细胞大量表达CSPGs;在AG1478组脊髓组织中CSPGs和GFAP的荧光强度均明显低于照组(P<0.01),这表明AG1478能明显抑制CSPGs和GFAP的表达。
     结论:脊髓损伤后星形胶质细胞明显上调pEGFR的表达,而表皮生长因子受体抑制剂AG1478通过对星形胶质细胞上pEGFR活化的抑制而导致其对脊髓损伤后反应性星形胶质增生及抑制因子CSPGs有明显抑制作用。
     第三部分表皮生长因子受体抑制剂AG1478对大鼠脊髓损伤后髓鞘脱失及髓鞘相关抑制因子表达的影响
     目的:外伤性脊髓损伤不仅造成神经元及轴突严重损伤而且导致少突胶质细胞受到严重伤害,而少突胶质细胞和轴突损害最终导致脊髓髓鞘的大量脱失。同时少突胶质细胞损伤后会释放大量髓鞘相关抑制分子,这些抑制分子对脊髓损伤后轴突的再生产生极其不利的影响。而表皮生长因子受体抑制剂AG1478对脊髓损伤有良好的治疗作用,故我们试图应用表皮生长因子受体抑制剂AG1478干预脊髓损伤以观察AG1478对大鼠脊髓损伤后髓鞘脱失及髓鞘相关抑制因子MAG、Nogo-A和OMgp mRNA表达的影响。
     方法:将33只健康雌性成年SD大鼠随机分为损伤对照组(重物坠落打击法建立脊髓损伤模型后立即在损伤局部给予含二甲基亚砜的明胶海绵外敷作对照治疗),AG1478治疗组(重物坠落打击法建立模型后立即在损伤局部给予含有0.2ml 2mM的AG1478的明胶海绵外敷治疗)和假手术组(只切除椎板并立即在损伤局部给予含二甲基亚砜的明胶海绵外敷作对照治疗)。各组大鼠均在脊髓损伤后14和28天处死。应用勒克司坚牢蓝染色(Luxol Fast Blue)方法分析各组大鼠脊髓损伤后28天髓鞘脱失情况。利用RT-PCR方法检测各组大鼠脊髓损伤后14天、28天髓鞘相关抑制因子MAG、Nogo-A和OMgp mRNA表达的情况。
     结果:在假手术组大鼠脊髓切片Luxol Fast Blue染色中,脊髓组织呈均匀的亮蓝色;而在对照组中损伤部位不能正常着色,这提示病灶中心髓鞘严重脱失;而在AG1478治疗组中脊髓组织切片不能正常着色的范围明显缩小,其髓鞘脱失面积明显小于对照组(P<0.01)。而髓鞘相关抑制分子MAG、Nogo-A和OMgp mRNA在假手术组有较弱的表达;在损伤对照组,脊髓损伤后14天及28天髓鞘相关抑制分子的表达均明显增加;而在AG1478组,脊髓损伤后14天及28天髓鞘相关抑制分子的表达均明显低于对照组(P<0.01)。
     结论:局部应用表皮生长因子受体抑制剂AG1478可改善大鼠脊髓损伤后髓鞘的脱失并能减少大鼠脊髓损伤后髓鞘相关抑制分子的MAG、Nogo-A和OMgp mRNA表达。
Part one The effects of epidermal growth factor receptor inhibitor AG1478 on GAP-43 expression and motor functional impairment after spinal cord injury in rats
     Objective The formation of glial scar and production of myelin-associated inhibitory molecules are two main obstacles to axonal regeneration after spinal cord injury. Recent studies have indicated that epidermal growth factor receptor (EGFR) activation triggered quiescent astrocytes into becoming reactive astrocytes and forming glial scar. And epidermal growth factor receptor inhibitors AG1478 promoted the axonal regeneration post optic injury by attenuating the inhibition effects of myelin-associated inhibitor molecule and chondroitin sulfate proteoglycans of axon. Therefore epidermal growth factor receptor inhibitors may attenuate the function impairment by promoting the injury axon regeneration after spinal cord injury. In these experiments we would administer the epidermal growth factor receptor inhibitors AG1478 in the local lesion and then observed whether AG1478 could upregulate the expression of growth associated protein-43(GAP-43) and attenuate the function impairment post injury.
     Methods The rats spinal cord injury model were built by using the weight-drop method, and sham-operated animals underwent T10 laminectomy alone.48 SD health female adult rats were randomly divided into 3 equal groups:sham group, injury control group and AG1478-treated group. The rats in AG1478-treated group were locally administered 0.2 ml 2mM AG1478 using a Gelfoam as a carrier, while the sham group and control group were locally administered dimethylsulfoxid also using a Gelfoam as a carrier. Immunofluorescence staining and Western blotting was used to detect the expression of growth associated protein-43 (GAP-43) 28 day post injury. The BBB scores of the hindlimb locomotor were collected at the first day post injury and weekly afterward (8 weeks). The body weight of all rats was measured previous injury and weekly afterward post injury (8 weeks).
     Results Immunofluorescence staining results showed that the positive labeling cells of GAP-43 in AG1478 group were more than that of injury control group (P< 0.01). Western blot results showed that the level of GAP-43 protein expression was significant higher in AG1478 group than that in the control group or sham group (P< 0.01). Hindlimb of rats in injury control group and AG1478-treated group were almost totally paralysed at first day post injury, which indicated that the two group rats underwent the same degree injury. Statistical analysis by repeated-measurement ANOVA indicated that the BBB scores of AG1478 group rats were significantly higher than that of control group rats. And statistical analysis by repeated-measurement ANOVA indicated that the body weight of AG1478 group rats were significantly higher than that of control group rats. (P<0.05).
     Conclusion Acute local administration of epidermal growth factory receptor inhibitors AG1478 in the contusion site could attenuate locomotor function impairment in spinal cord injured rats and significantly upregulated the level of GAP-43 expression. The robust effects of epidermal growth factor receptor inhibitors AG1478 on spinal cord injury indicated that epidermal growth factor receptor inhibitors could be efficient protective agent on central nervous system injury.
     Part two Effects of epidermal growth factor receptor inhibitor AG1478 on reactive astrogliosis after spinal cord injury in rats
     Objective Astrogliosis are thought to play a key role in the pathophysiology of spinal cord injury (SCI), and there are a close relationship between astrogliosis and the activation of epidermal growth factor receptor in the pathophysiology of central nervous system injury. We sought to administer epidermal growth factor recepter AG1478 to treat spinal cord injury in order to investigate the effects of epidermal growth factor receptor inhibitor AG1478 on the expression of pEGFR in astrocytes and reactive astrogliosis and production of axon growth inhibitory factor chondroitin sulphate proteoglycans after rat spinal cord injury.
     Methods The rats spinal cord injury model were built by using the weight-drop method, and sham-operated animals underwent T10 laminectomy alone. Forty five SD health female adult rats were randomly divided into 3 equal groups:sham group, injury control group and AG1478-treated group. The rats in AG1478-treated group were locally administered 0.2 ml 2mM AG 1478 using a Gelfoam as a carrier, while the sham group and control group were locally administered dimethylsulfoxid also using a Gelfoam as a carrier. All rats were sacrificed at 14d and 28d. The expression of GFAP at day 14 and day 28 after spinal cord injury were detected by immunofluorescence and western blot. The expression of phosphorylated epidermal growth factor receptor inhibitors in astrocytes at day 14 after spinal cord injury was observed by double-immunofluorescence and the expression of chondroitin sulphate proteoglycans in astrocytes at day 28 after spinal cord injury were observed by double-immunofluorescence.
     Results GFAP-positive cells were sparse and slim in the sham group. In contrast, activated astrocytes characterized by the hypertrophy of astrocytic processes and up-regulation of GFAP were present at 2 weeks after SCI and the typical glial scar became evident at 4 weeks after SCI in the control group. While the up-regulation GFAP was significantly suppressed at 2 weeks after SCI and the dense glial scar was not observed at 4 weeks after SCI in the AG1478 group. Western blot analysis also showed that the GFAP expression of AG1478-treated group at 2 and 4 weeks after SCI was significant lower than that of control group (P< 0.01). By double-immunofluorescence, positive labeling of pEGFR and GFAP was weakly present or absent in sham group and no colozalition of pEGFR and GFAP appeared in astrocytes in sham group, which indicated that the astrocytes in sham group did not expressed pEGFR. While positive labeling of pEGFR and GFAP was strongly present in control group. The cyst and glial scar were formed at the lesion center in control group, and there was significant colozalition of pEGFR and GFAP in the glia scar around the cyst. This indicated that astrocytes after spinal cord injury expressed pEGFR. The immunofluorescence intensity of pEGFR and GFAP in AG1478-treated group was significantly weaker compared with that in control group(P<0.01), which indicated that the expression of pEGFR and GFAP was suppressed by AG1478. No colozalition of pEGFR and GFAP appeared in AG1478-treated group. Positive labeling of CSPGs and GFAP was weakly present in sham group, while positive labeling of CSPGs and GFAP was strongly present in control group. Moreover, there was significant colozalition of CSPGs and GFAP in the glial scar, which indicated that active astrocytes significanlty expressed CSPGs. The immunofluorescence intensity of CSPGs and GFAP in AG1478-treated group was significantly weaker compared with that in control group(P<0.01), which indicated that the expression of CSPGs and GFAP was suppressed by AG1478.
     Conclusion Astrocytes were significantly up-regulated the expression of pEGFR after SCI. Epidermal growth factor receptor inhibitor AG1478 can attenuate the reactive astrogliosis and the prodution of inhibitory factor CSPGs after SCI by inhibiting the active pEGFR in astrocytes after spinal cord injury.
     Part three Effects of epidermal growth factor receptor inhibitor AG1478 on demyelination and the expression of myelin-associated inhibitory molecules after spinal cord injury in rats
     Objective Traumatic spinal cord injury (SCI) not only cause damage to neuron and axon but also induces harmful effects on the oligodendrocytes, which contribute to severe demyelination. At the same time, the damage oligodendrocytes release lots of myelin-associated inhibitory molecules, which induce an harmful influence on the axonal degeneration after spinal cord injury. Since epidermal growth factor receptor inhibitor AG1478 has a robust effects on the function recovery of spinal cord injury, we sought to administrate AG1478 to treat spinal cord injury in order to investigate the effects of AG1478 on demyelination and the mRNA expression of myelin-associated inhibitory molecules MAG、Nogo-A and OMgp after rat spinal cord injury.
     Methods Thirty three health female adult SD rats were randomly divided into 3 equal groups:control group undergoing SCI by weight drop method and acute local administration dimethylsulfoxide using a Gelfoam as a carrier after SCI, AG1478-treated group undergoing SCI by weight drop method and acute local administration 0.2 ml 2mM AG1478 using a Gelfoam as a carrier after SCI, and sham group undergoing T10 laminectomy alone and acute local administration dimethylsulfoxide using a Gelfoam as a carrier postoperation. All rats were sacrificed at 14d and 28d post injury. The demyelination of all group rats at day 28 after spinal cord injury was observed by Luxol Fast Blue staining. The mRNA expression of MAG、Nogo-A and OMgp in all group rats at day 14 and day 28 after spinal cord injury were detected by RT-PCR.
     Result By Luxol Fast Blue staining, sham-operated spinal cord tissue was characterized by light blue equably across the entire spinal cords, while there was normal coloring deletion area in the lesion center of injury spinal cord tissue, which indicated that demyelination was severe in the lesion center. And the normal coloring deletion area was significantly reduced in the lesion center of AG1478-treated group spinal cord tissues. The demyelination area of AG1478-treated group was significant smaller compared with that of control group(P<0.01). The mRNA expression of myelin-associated inhibitory molecules MAG、Nogo-A and OMgp of sham group was weak post injury, while the mRNA expression of myelin-associated inhibitory molecules MAG、Nogo-A and OMgp of control group significantly increased at day 14 and day 28 after spinal cord injury. The mRNA expression level of myelin-associated inhibitory molecules MAG、Nogo-A and OMgp of AG1478-treated group was significant lower compared to that of control group (P<0.01).
     Conclusion Local administration of epidermal growth factor receptor inhibitor AG1478 can attenuate the demyelination after spinal cord injury and reduces the mRNA expression level of myelin-associated inhibitory molecules MAG、Nogo-A and OMgp post injury.
引文
[1]Tator CH, Fehlings MG Review of the secondary injury theory of acute spinal cord trauma with emphasis on vascular mechanisms. J Neurosurg 1991;75 (1):15-26.
    [2]Hagg T, Oudega M. Degenerative and spontaneous regenerative processes after spinal cord injury. J Neurotrauma 2006;23 (3-4):264-280.
    [3]Ottomanelli L, Lind L. Review of critical factors related to employment after spinal cord injury:implications for research and vocational services. J Spinal Cord Med 2009;32 (5):503-531.
    [4]Erschbamer M, Pemold K, Olson L. Inhibiting epidermal growth factor receptor improves structural, locomotor, sensory, and bladder recovery from experimental spinal cord injury. J Neurosci 2007;27 (24):6428-6435.
    [5]le Gros Clark WE. The problem of neuronal regeneration in the central nervous system:Ⅱ. The insertion of peripheral nerve stumps into the brain. J Anat 1943;77 (Pt 3):251-259.
    [6]Harel NY, Strittmatter SM. Can regenerating axons recapitulate developmental guidance during recovery from spinal cord injury? Nat Rev Neurosci 2006;7 (8):603-616.
    [7]Gwak YS, Nam TS, Paik KS, et al. Attenuation of mechanical hyperalgesia following spinal cord injury by administration of antibodies to nerve growth factor in the rat. Neurosci Lett 2003;336 (2):117-120.
    [8]Grimpe B, Silver J. The extracellular matrix in axon regeneration. Prog Brain Res 2002;137:333-349.
    [9]Taylor L, Jones L, Tuszynski MH, et al. Neurotrophin-3 gradients established by lentiviral gene delivery promote short-distance axonal bridging beyond cellular grafts in the injured spinal cord. J Neurosci 2006;26 (38):9713-9721.
    [10]Freund P, Schmidlin E, Wannier T, et al. Nogo-A-specific antibody treatment enhances sprouting and functional recovery after cervical lesion in adult primates. Nat Med 2006; 12 (7):790-792.
    [11]Fitch MT, Silver J. CNS injury, glial scars, and inflammation:Inhibitory extracellular matrices and regeneration failure. Exp Neurol 2008;209 (2):294-301.
    [12]Chaudhry N, Filbin MT. Myelin-associated inhibitory signaling and strategies to overcome inhibition. J Cereb Blood Flow Metab 2007;27 (6):1096-1107.
    [13]Giger RJ, Venkatesh K, Chivatakarn O, et al. Mechanisms of CNS myelin inhibition:evidence for distinct and neuronal cell type specific receptor systems. Restor Neurol Neurosci 2008;26 (2-3):97-115.
    [14]Kubo T, Yamashita T. Rho-ROCK inhibitors for the treatment of CNS injury. Recent Pat CNS Drug Discov 2007;2 (3):173-179.
    [15]Kastin AJ, Pan W. Targeting neurite growth inhibitors to induce CNS regeneration. Curr Pharm Des 2005;11 (10):1247-1253.
    [16]Ruff RL, McKerracher L, Selzer ME. Repair and neurorehabilitation strategies for spinal cord injury. Ann N Y Acad Sci 2008; 1142:1-20.
    [17]Koprivica V, Cho KS, Park JB, et al. EGFR activation mediates inhibition of axon regeneration by myelin and chondroitin sulfate proteoglycans. Science 2005;310(5745):106-110.
    [18]Donnelly DJ, Popovich PG Inflammation and its role in neuroprotection, axonal regeneration and functional recovery after spinal cord injury. Exp Neurol 2008;209 (2):378-388.
    [19]Tian DS, Xie MJ, Yu ZY, et al. Cell cycle inhibition attenuates microglia induced inflammatory response and alleviates neuronal cell death after spinal cord injury in rats. Brain Res 2007;1135 (1):177-185.
    [20]Tian DS, Dong Q, Pan DJ, et al. Attenuation of astrogliosis by suppressing of microglial proliferation with the cell cycle inhibitor olomoucine in rat spinal cord injury model. Brain Res 2007; 1154:206-214.
    [21]Tian DS, Yu ZY, Xie MJ, et al. Suppression of astroglial scar formation and enhanced axonal regeneration associated with functional recovery in a spinal cord injury rat model by the cell cycle inhibitor olomoucine. J Neurosci Res 2006;84(5):1053-1063.
    [22]Smith GM, Strunz C. Growth factor and cytokine regulation of chondroitin sulfate proteoglycans by astrocytes. Glia 2005;52 (3):209-218.
    [23]Hwang SY, Jung JS, Kim TH, et al. Ionizing radiation induces astrocyte gliosis through microglia activation. Neurobiol Dis 2006;21 (3):457-467.
    [24]Rohl C, Lucius R, Sievers J. The effect of activated microglia on astrogliosis parameters in astrocyte cultures. Brain Res 2007;1129 (1):43-52.
    [25]Moynagh PN. The interleukin-1 signalling pathway in astrocytes:a key contributor to inflammation in the brain. J Anat 2005;207 (3):265-269.
    [26]Liu B, Neufeld AH. Activation of epidermal growth factor receptor causes astrocytes to form cribriform structures. Glia 2004;46 (2):153-168.
    [27]Holley JE, Gveric D, Newcombe J, et al. Astrocyte characterization in the multiple sclerosis glial scar. Neuropathol Appl Neurobiol 2003;29 (5):434-444.
    [28]Ferrer I, Alcantara S, Ballabriga J, et al. Transforming growth factor-alpha (TGF-alpha) and epidermal growth factor-receptor (EGF-R) immunoreactivity in normal and pathologic brain. Prog Neurobiol 1996;49 (2):99-123.
    [29]Jin K, Mao XO, Sun Y, et al. Heparin-binding epidermal growth factor-like growth factor:hypoxia-inducible expression in vitro and stimulation of neurogenesis in vitro and in vivo. J Neurosci 2002;22 (13):5365-5373.
    [30]Hayashi Y, Yamashita J, Watanabe T. Molecular genetic analysis of deep-seated glioblastomas. Cancer Genet Cytogenet 2004;153 (1):64-68.
    [31]Liu B, Chen H, Johns TG, et al. Epidermal growth factor receptor activation: an upstream signal for transition of quiescent astrocytes into reactive astrocytes after neural injury. J Neurosci 2006;26 (28):7532-7540.
    [32]Planas AM, Justicia C, Soriano MA, et al. Epidermal growth factor receptor in proliferating reactive glia following transient focal ischemia in the rat brain. Glia 1998;23 (2):120-129.
    [33]Schachtrup C, Lu P, Jones LL, et al. Fibrinogen inhibits neurite outgrowth via beta 3 integrin-mediated phosphorylation of the EGF receptor. Proc Natl Acad Sci U S A 2007;104 (28):11814-11819.
    [34]Mosesson MW. Fibrinogen and fibrin structure and functions. J Thromb Haemost 2005;3 (8):1894-1904.
    [35]Schwab JM, Tuli SK, Failli V. The Nogo receptor complex:confining molecules to molecular mechanisms. Trends Mol Med 2006;12 (7):293-297.
    [36]Liu B, Neufeld AH. Activation of epidermal growth factor receptor signals induction of nitric oxide synthase-2 in human optic nerve head astrocytes in glaucomatous optic neuropathy. Neurobiol Dis 2003; 13 (2):109-123.
    [37]Pautier P, Joly F, Kerbrat P, et al. Phase II study of gefitinib in combination with paclitaxel (P) and carboplatin (C) as second-line therapy for ovarian, tubal or peritoneal adenocarcinoma (1839IL/0074). Gynecol Oncol; 116 (2):157-162.
    [1]David S, Aguayo AJ. Axonal elongation into peripheral nervous system "bridges" after central nervous system injury in adult rats. Science 1981;214 (4523):931-933.
    [2]Tian DS, Liu JL, Xie MJ, et al. Tamoxifen attenuates inflammatory-mediated damage and improves functional outcome after spinal cord injury in rats. J Neurochem 2009;109 (6):1658-1667.
    [3]Kastin AJ, Pan W. Targeting neurite growth inhibitors to induce CNS regeneration. Curr Pharm Des 2005;11 (10):1247-1253.
    [4]Ruff RL, McKerracher L, Selzer ME. Repair and neurorehabilitation strategies for spinal cord injury. Ann N Y Acad Sci 2008;1142:1-20.
    [5]Tian DS, Yu ZY, Xie MJ, et al. Suppression of astroglial scar formation and enhanced axonal regeneration associated with functional recovery in a spinal
    cord injury rat model by the cell cycle inhibitor olomoucine. J Neurosci Res 2006;84(5):1053-1063.
    [6]Tang X, Davies JE, Davies SJ. Changes in distribution, cell associations, and protein expression levels of NG2, neurocan, phosphacan, brevican, versican V2, and tenascin-C during acute to chronic maturation of spinal cord scar tissue. J Neurosci Res 2003;71 (3):427-444.
    [7]Kubo T, Yamashita T. Rho-ROCK inhibitors for the treatment of CNS injury. Recent Pat CNS Drug Discov 2007;2 (3):173-179.
    [8]Planas AM, Justicia C, Soriano MA, et al. Epidermal growth factor receptor in proliferating reactive glia following transient focal ischemia in the rat brain. Glia 1998;23(2):120-129.
    [9]Jin K, Mao XO, Sun Y, et al. Heparin-binding epidermal growth factor-like growth factor:hypoxia-inducible expression in vitro and stimulation of neurogenesis in vitro and in vivo. J Neurosci 2002;22 (13):5365-5373.
    [10]Ferrer I, Alcantara S, Ballabriga J, et al. Transforming growth factor-alpha (TGF-alpha) and epidermal growth factor-receptor (EGF-R) immunoreactivity in normal and pathologic brain. Prog Neurobiol 1996;49 (2):99-123.
    [11]Rabchevsky AG, Weinitz JM, Coulpier M, et al. A role for transforming growth factor alpha as an inducer of astrogliosis. J Neurosci 1998; 18 (24):10541-10552.
    [12]Liu L, Cash TP, Jones RG, et al. Hypoxia-induced energy stress regulates mRNA translation and cell growth. Mol Cell 2006;21 (4):521-531.
    [13]Kang MK, Kang SK. Interleukin-6 induces proliferation in adult spinal cord-derived neural progenitors via the JAK2/STAT3 pathway with EGF-induced MAPK phosphorylation. Cell Prolif 2008;41 (3):377-392.
    [14]Koprivica V, Cho KS, Park JB, et al. EGFR activation mediates inhibition of axon regeneration by myelin and chondroitin sulfate proteoglycans. Science 2005;310(5745):106-110.
    [15]Gruner JA. A monitored contusion model of spinal cord injury in the rat. J Neurotrauma 1992;9 (2):123-126; discussion 126-128.
    [16]Basso DM, Beattie MS, Bresnahan JC. A sensitive and reliable locomotor rating scale for open field testing in rats. J Neurotrauma 1995;12 (1):1-21.
    [17]Labombarda F, Gonzalez SL, Gonzalez DM, et al. Cellular basis for progesterone neuroprotection in the injured spinal cord. J Neurotrauma 2002;19(3):343-355.
    [18]Tator CH. Update on the pathophysiology and pathology of acute spinal cord injury. Brain Pathol 1995;5 (4):407-413.
    [19]Shibuya S, Miyamoto O, Itano T, et al. Temporal progressive antigen expression in radial glia after contusive spinal cord injury in adult rats. Glia 2003;42(2):172-183.
    [20]Guth L, Zhang Z, Steward O. The unique histopathological responses of the injured spinal cord. Implications for neuroprotective therapy. Ann N Y Acad Sci 1999;890:366-384.
    [21]Jones LL, Margolis RU, Tuszynski MH. The chondroitin sulfate proteoglycans neurocan, brevican, phosphacan, and versican are differentially regulated following spinal cord injury. Exp Neural 2003;182 (2):399-411.
    [22]Morgenstern DA, Asher RA, Fawcett JW. Chondroitin sulphate proteoglycans in the CNS injury response. Prog Brain Res 2002;137:313-332.
    [23]Vourc'h P, Andres C. Oligodendrocyte myelin glycoprotein (OMgp):evolution, structure and function. Brain Res Brain Res Rev 2004;45 (2):115-124.
    [24]Oertle T, van der Haar ME, Bandtlow CE, et al. Nogo-A inhibits neurite outgrowth and cell spreading with three discrete regions. J Neurosci 2003;23 (13):5393-5406.
    [25]Domeniconi M, Cao Z, Spencer T, et al. Myelin-associated glycoprotein interacts with the Nogo66 receptor to inhibit neurite outgrowth. Neuron 2002;35 (2):283-290.
    [26]Schachtrup C, Lu P, Jones LL, et al. Fibrinogen inhibits neurite outgrowth via beta 3 integrin-mediated phosphorylation of the EGF receptor. Proc Natl Acad Sci U S A 2007;104(28):11814-11819.
    [27]Liu B, Neufeld AH. Activation of epidermal growth factor receptors in astrocytes:from development to neural injury. J Neurosci Res 2007;85 (16):3523-3529.
    [28]Rio C, Perez-Cerda F, Matute C, et al. Preparation of a monoclonal antibody to a glycidic epitope of the epidermal growth factor receptor that recognizes inhibitors of astrocyte proliferation and reactive microglia. J Neurosci Res 1995;40(6):776-786.
    [29]Liu B, Chen H, Johns TG, et al. Epidermal growth factor receptor activation: an upstream signal for transition of quiescent astrocytes into reactive astrocytes after neural injury. J Neurosci 2006;26 (28):7532-7540.
    [30]Rivlin AS, Tator CH. Objective clinical assessment of motor function after experimental spinal cord injury in the rat. J Neurosurg 1977;47 (4):577-581.
    [31]Gale K, Kerasidis H, Wrathall JR. Spinal cord contusion in the rat:behavioral analysis of functional neurologic impairment. Exp Neurol 1985;88 (1):123-134.
    [32]Kerasidis H, Wrathall JR, Gale K. Behavioral assessment of functional deficit in rats with contusive spinal cord injury. J Neurosci Methods 1987;20 (2):167-179.
    [33]Skene JH, Willard M. Electrophoretic analysis of axonally transported proteins in toad retinal ganglion cells. J Neurochem 1981;37 (1):79-87.
    [34]Gorgels TG, Van Lookeren Campagne M, Oestreicher AB, et al. B-50/GAP43 is localized at the cytoplasmic side of the plasma membrane in developing and adult rat pyramidal tract. J Neurosci 1989;9 (11):3861-3869.
    [35]Zhu Q, Julien JP. A key role for GAP-43 in the retinotectal topographic organization. Exp Neurol 1999; 155 (2):228-242.
    [36]Dubroff JG, Stevens RT, Hitt J, et al. Anomalous functional organization of barrel cortex in GAP-43 deficient mice. Neuroimage 2006;29 (4):1040-1048.
    [37]Regland B, Lehmann W, Abedini I, et al. Treatment of Alzheimer's disease with clioquinol. Dement Geriatr Cogn Disord 2001;12 (6):408-414.
    [38]Schmitt AB, Breuer S, Voell M, et al. GAP-43 (B-50) and C-Jun are up-regulated in axotomized neurons of Clarke's nucleus after spinal cord injury in the adult rat. Neurobiol Dis 1999;6 (2):122-130.
    [39]Wang C, Zhang YF, Huang XY. Effects of continuous sciatic nerve block by tetrodotoxin on growth associated protein-43 expression in dorsal root ganglions of normal and sciatic nerve injury rats. Chin Med Sci J 2007;22 (4):264.
    [40]Mosesson MW. Fibrinogen and fibrin structure and functions. J Thromb Haemost 2005;3 (8):1894-1904.
    [41]Donnelly DJ, Popovich PG Inflammation and its role in neuroprotection, axonal regeneration and functional recovery after spinal cord injury. Exp Neurol 2008;209 (2):378-388.
    [42]Erschbamer M, Pernold K, Olson L. Inhibiting epidermal growth factor receptor improves structural, locomotor, sensory, and bladder recovery from experimental spinal cord injury. J Neurosci 2007;27 (24):6428-6435.
    [1]Tian DS, Yu ZY, Xie MJ, et al. Suppression of astroglial scar formation and enhanced axonal regeneration associated with functional recovery in a spinal cord injury rat model by the cell cycle inhibitor olomoucine. J Neurosci Res 2006;84(5):1053-1063.
    [2]Windle WF, Chambers WW. Regeneration in the spinal cord of the cat and dog. AMA Arch Neurol Psychiatry 1951;65 (2):261-262.
    [3]Fitch MT, Silver J. CNS injury, glial scars, and inflammation:Inhibitory extracellular matrices and regeneration failure. Exp Neurol 2008;209 (2):294-301.
    [4]Shields LB, Zhang YP, Burke DA, et al. Benefit of chondroitinase ABC on sensory axon regeneration in a laceration model of spinal cord injury in the rat. Surg Neurol 2008;69 (6):568-577; discussion 577.
    [5]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. Eur J Neurosci 2001;13 (2):257-268.
    [6]Bandtlow C, Zachleder T, Schwab ME. Oligodendrocytes arrest neurite growth by contact inhibition. J Neurosci 1990;10 (12):3837-3848.
    [7]Fawcett JW, Fersht N, Housden L, et al. Axonal growth on astrocytes is not inhibited by oligodendrocytes. J Cell Sci 1992;103 (Pt 2):571-579.
    [8]Fidler PS, Schuette K, Asher RA, et al. Comparing astrocytic cell lines that are inhibitory or permissive for axon growth:the major axon-inhibitory proteoglycan is NG2. J Neurosci 1999;19 (20):8778-8788.
    [9]Smith GM, Strunz C. Growth factor and cytokine regulation of chondroitin sulfate proteoglycans by astrocytes. Glia 2005;52 (3):209-218.
    [10]Liu B, Neufeld AH. Activation of epidermal growth factor receptor causes astrocytes to form cribriform structures. Glia 2004;46 (2):153-168.
    [11]Liu B, Neufeld AH. Activation of epidermal growth factor receptor signals induction of nitric oxide synthase-2 in human optic nerve head astrocytes in glaucomatous optic neuropathy. Neurobiol Dis 2003;13 (2):109-123.
    [12]Holley JE, Gveric D, Newcombe J, et al. Astrocyte characterization in the multiple sclerosis glial scar. Neuropathol Appl Neurobiol 2003;29 (5):434-444.
    [13]Erschbamer M, Pernold K, Olson L. Inhibiting epidermal growth factor receptor improves structural, locomotor, sensory, and bladder recovery from experimental spinal cord injury. J Neurosci 2007;27 (24):6428-6435.
    [14]le Gros Clark WE. The problem of neuronal regeneration in the central nervous system:Ⅱ. The insertion of peripheral nerve stumps into the brain. J Anat 1943;77(Pt 3):251-259.
    [15]Freund P, Schmidlin E, Wannier T, et al. Nogo-A-specific antibody treatment enhances sprouting and functional recovery after cervical lesion in adult primates. Nat Med 2006; 12 (7):790-792.
    [16]Goshgarian HG. The crossed phrenic phenomenon:a model for plasticity in the respiratory pathways following spinal cord injury. J Appl Physiol 2003;94 (2):795-810.
    [17]Li Y, Raisman G. Schwann cells induce sprouting in motor and sensory axons in the adult rat spinal cord. J Neurosci 1994;14 (7):4050-4063.
    [18]Taylor L, Jones L, Tuszynski MH, et al. Neurotrophin-3 gradients established by lentiviral gene delivery promote short-distance axonal bridging beyond
    cellular grafts in the injured spinal cord. J Neurosci 2006;26 (38):9713-9721.
    [19]Guth L, Barrett CP, Donati EJ, et al. Essentiality of a specific cellular terrain for growth of axons into a spinal cord lesion. Exp Neurol 1985;88 (1):1-12.
    [20]Iseda T, Okuda T, Kane-Goldsmith N, et al. Single, high-dose intraspinal injection of chondroitinase reduces glycosaminoglycans in injured spinal cord and promotes corticospinal axonal regrowth after hemisection but not contusion. J Neurotrauma 2008;25 (4):334-349.
    [21]Iseda T, Nishio T, Kawaguchi S, et al. Spontaneous regeneration of the corticospinal tract after transection in young rats:a key role of reactive astrocytes in making favorable and unfavorable conditions for regeneration. Neuroscience 2004;126 (2):365-374.
    [22]Roonprapunt C, Huang W, Grill R, et al. Soluble cell adhesion molecule Ll-Fc promotes locomotor recovery in rats after spinal cord injury. J Neurotrauma 2003;20 (9):871-882.
    [23]Jin K, Mao XO, Sun Y, et al. Heparin-binding epidermal growth factor-like growth factor:hypoxia-inducible expression in vitro and stimulation of neurogenesis in vitro and in vivo. J Neurosci 2002;22 (13):5365-5373.
    [24]Usul H, Cakir E, Cobanoglu U, et al. The effects of tyrphostine Ag 556 on experimental spinal cord ischemia reperfusion injury. Surg Neurol 2004;61 (1):45-54; discussion 54.
    [25]Profyris C, Cheema SS, Zang D, et al. Degenerative and regenerative mechanisms governing spinal cord injury. Neurobiol Dis 2004; 15 (3):415-436.
    [26]Kigerl KA, McGaughy VM, Popovich PG. Comparative analysis of lesion development and intraspinal inflammation in four strains of mice following spinal contusion injury. J Comp Neurol 2006;494 (4):578-594.
    [27]Sugawara T, Lewen A, Gasche Y, et al. Overexpression of SOD1 protects vulnerable motor neurons after spinal cord injury by attenuating mitochondrial cytochrome c release. Faseb J 2002;16 (14):1997-1999.
    [28]Matyja E, Naganska E, Taraszewska A, et al. The mode of spinal motor neurons degeneration in a model of slow glutamate excitotoxicity in vitro. Folia Neuropathol 2005;43 (1):7-13.
    [29]Faulkner JR, Herrmann JE, Woo MJ, et al. Reactive astrocytes protect tissue and preserve function after spinal cord injury. J Neurosci 2004;24 (9):2143-2155.
    [30]Planas AM, Justicia C, Soriano MA, et al. Epidermal growth factor receptor in proliferating reactive glia following transient focal ischemia in the rat brain. Glia 1998;23 (2):120-129.
    [31]Morino T, Ogata T, Horiuchi H, et al. Delayed neuronal damage related to microglia proliferation after mild spinal cord compression injury. Neurosci Res 2003;46(3):309-318.
    [32]Gibbons HM, Dragunow M. Microglia induce neural cell death via a proximity-dependent mechanism involving nitric oxide. Brain Res 2006; 1084 (1):1-15.
    [33]Tian DS, Xie MJ, Yu ZY, et al. Cell cycle inhibition attenuates microglia induced inflammatory response and alleviates neuronal cell death after spinal cord injury in rats. Brain Res 2007;1135 (1):177-185.
    [34]Tian DS, Dong Q, Pan DJ, et al. Attenuation of astrogliosis by suppressing of microglial proliferation with the cell cycle inhibitor olomoucine in rat spinal cord injury model. Brain Res 2007; 1154:206-214.
    [35]Balasingam V, Tejada-Berges T, Wright E, et al. Reactive astrogliosis in the neonatal mouse brain and its modulation by cytokines. J Neurosci 1994; 14 (2):846-856.
    [36]Hwang SY, Jung JS, Kim TH, et al. Ionizing radiation induces astrocyte gliosis through microglia activation. Neurobiol Dis 2006;21 (3):457-467.
    [37]Kordek R, Nerurkar VR, Liberski PP, et al. Heightened expression of tumor necrosis factor alpha, interleukin 1 alpha, and glial fibrillary acidic protein in experimental Creutzfeldt-Jakob disease in mice. Proc Natl Acad Sci U S A 1996;93(18):9754-9758.
    [38]Moynagh PN. The interleukin-1 signalling pathway in astrocytes:a key contributor to inflammation in the brain. J Anat 2005;207 (3):265-269.
    [39]Walz W. Role of glial cells in the regulation of the brain ion microenvironment. Prog Neurobiol 1989;33 (4):309-333.
    [40]Massey JM, Hubscher CH, Wagoner MR, et al. Chondroitinase ABC digestion of the perineuronal net promotes functional collateral sprouting in the cuneate nucleus after cervical spinal cord injury. J Neurosci 2006;26 (16):4406-4414.
    [41]Canning DR, Hoke A, Malemud CJ, et al. A potent inhibitor of neurite outgrowth that predominates in the extracellular matrix of reactive astrocytes. Int J Dev Neurosci 1996; 14 (3):153-175.
    [42]Tacconi MT. Neuronal death:is there a role for astrocytes? Neurochem Res 1998;23 (5):759-765.
    [43]Mrak RE, Griffin WS. Interleukin-1, neuroinflammation, and Alzheimer's disease. Neurobiol Aging 2001;22 (6):903-908.
    [44]Duncan AJ, Heales SJ. Nitric oxide and neurological disorders. Mol Aspects Med 2005;26(1-2):67-96.
    [45]Davies JE, Tang X, Denning JW, et al. Decorin suppresses neurocan, brevican, phosphacan and NG2 expression and promotes axon growth across adult rat spinal cord injuries. Eur J Neurosci 2004; 19 (5):1226-1242.
    [46]Klapka N, Hermanns S, Straten G, et al. Suppression of fibrous scarring in spinal cord injury of rat promotes long-distance regeneration of corticospinal tract axons, rescue of primary motoneurons in somatosensory cortex and significant functional recovery. Eur J Neurosci 2005;22 (12):3047-3058.
    [47]Zhang H, Uchimura K, Kadomatsu K. Brain keratan sulfate and glial scar formation. Ann N Y Acad Sci 2006; 1086:81-90.
    [48]Niederost BP, Zimmermann DR, Schwab ME, et al. Bovine CNS myelin contains neurite growth-inhibitory activity associated with chondroitin sulfate proteoglycans. J Neurosci 1999;19 (20):8979-8989.
    [49]Kubo T, Yamashita T. Rho-ROCK inhibitors for the treatment of CNS injury. Recent Pat CNS Drug Discov 2007;2 (3):173-179.
    [50]Liu B, Chen H, Johns TG, et al. Epidermal growth factor receptor activation: an upstream signal for transition of quiescent astrocytes into reactive astrocytes after neural injury. J Neurosci 2006;26 (28):7532-7540.
    [51]Citri A, Yarden Y. EGF-ERBB signalling:towards the systems level. Nat Rev Mol Cell Biol 2006;7 (7):505-516.
    [52]Voldborg BR, Damstrup L, Spang-Thomsen M, et al. Epidermal growth factor receptor (EGFR) and EGFR mutations, function and possible role in clinical trials. Ann Oncol 1997;8 (12):1197-1206.
    [53]Gomez-Pinilla F, Knauer DJ, Nieto-Sampedro M. Epidermal growth factor receptor immunoreactivity in rat brain. Development and cellular localization. Brain Res 1988;438 (1-2):385-390.
    [54]Birecree E, Whetsell WO, Jr., Stoscheck C, et al. Immunoreactive epidermal growth factor receptors in neuritic plaques from patients with Alzheimer's disease. J Neuropathol Exp Neurol 1988;47 (5):549-560.
    [55]Hayashi Y, Yamashita J, Watanabe T. Molecular genetic analysis of deep-seated glioblastomas. Cancer Genet Cytogenet 2004; 153 (1):64-68.
    [56]Asher RA, Morgenstern DA, Fidler PS, et al. Neurocan is upregulated in injured brain and in cytokine-treated astrocytes. J Neurosci 2000;20 (7):2427-2438.
    [57]Asher RA, Morgenstern DA, Shearer MC, et al. Versican is upregulated in CNS injury and is a product of oligodendrocyte lineage cells. J Neurosci 2002;22 (6):2225-2236.
    [58]Jones LL, Yamaguchi Y, Stallcup WB, et al. NG2 is a major chondroitin sulfate proteoglycan produced after spinal cord injury and is expressed by macrophages and oligodendrocyte progenitors. J Neurosci 2002;22 (7):2792-2803.
    [59]Jones LL, Margolis RU, Tuszynski MH. The chondroitin sulfate proteoglycans neurocan, brevican, phosphacan, and versican are differentially regulated following spinal cord injury. Exp Neurol 2003;182 (2):399-411.
    [60]Jones LL, Sajed D, Tuszynski MH. Axonal regeneration through regions of chondroitin sulfate proteoglycan deposition after spinal cord injury:a balance of permissiveness and inhibition. J Neurosci 2003;23 (28):9276-9288.
    [61]Koprivica V, Cho KS, Park JB, et al. EGFR activation mediates inhibition of axon regeneration by myelin and chondroitin sulfate proteoglycans. Science 2005;310 (5745):106-110.
    [62]McKeon RJ, Jurynec MJ, Buck CR. The chondroitin sulfate proteoglycans neurocan and phosphacan are expressed by reactive astrocytes in the chronic CNS glial scar. J Neurosci 1999;19 (24):10778-10788.
    [63]Snow DM, Lemmon V, Carrino DA, et al. Sulfated proteoglycans in astroglial barriers inhibit neurite outgrowth in vitro. Exp Neurol 1990; 109 (1):111-130.
    [64]Kaneko M, Kubo T, Hata K, et al. Repulsion of cerebellar granule neurons by chondroitin sulfate proteoglycans is mediated by MAPK pathway. Neurosci Lett 2007;423(1):62-67.
    [1]Sahni V, Mukhopadhyay A, Tysseling V, et al. BMPRla and BMPRlb signaling exert opposing effects on gliosis after spinal cord injury. J Neurosci;30 (5):1839-1855.
    [2]Busch SA, Silver J. The role of extracellular matrix in CNS regeneration. Curr Opin Neurobiol 2007;17 (1):120-127.
    [3]Yiu G, He Z. Glial inhibition of CNS axon regeneration. Nat Rev Neurosci 2006;7 (8):617-627.
    [4]David S, Lacroix S. Molecular approaches to spinal cord repair. Annu Rev Neurosci 2003;26:411-440.
    [5]Tian DS, Liu JL, Xie MJ, et al. Tamoxifen attenuates inflammatory-mediated damage and improves functional outcome after spinal cord injury in rats. J Neurochem 2009;109 (6):1658-1667.
    [6]Becker D, Sadowsky CL, McDonald JW. Restoring function after spinal cord injury. Neurologist 2003;9 (1):1-15.
    [7]Liu XZ, Xu XM, Hu R, et al. Neuronal and glial apoptosis after traumatic spinal cord injury. J Neurosci 1997;17 (14):5395-5406.
    [8]McDonald JW, Levine JM, Qu Y. Multiple classes of the oligodendrocyte lineage are highly vulnerable to excitotoxicity. Neuroreport 1998;9 (12):2757-2762.
    [9]de Wit J, Verhaagen J. Role of semaphorins in the adult nervous system. Prog Neurobiol 2003;71 (2-3):249-267.
    [10]Grigoriadis N, Ben-Hur T, Karussis D, et al. Axonal damage in multiple sclerosis:a complex issue in a complex disease. Clin Neurol Neurosurg 2004;106(3):211-217.
    [11]Ludwin SK. Central nervous system remyelination:studies in chronically damaged tissue. Ann Neurol 1994;36 Suppl:S143-145.
    [12]Goldberg JL, Klassen MP, Hua Y, et al. Amacrine-signaled loss of intrinsic axon growth ability by retinal ganglion cells. Science 2002;296 (5574):1860-1864.
    [13]Kubo T, Yamashita T. Rho-ROCK inhibitors for the treatment of CNS injury. Recent Pat CNS Drug Discov 2007;2 (3):173-179.
    [14]Oertle T, van der Haar ME, Bandtlow CE, et al. Nogo-A inhibits neurite outgrowth and cell spreading with three discrete regions. J Neurosci 2003;23 (13):5393-5406.
    [15]Liu BP, Fournier A, GrandPre T, et al. Myelin-associated glycoprotein as a functional ligand for the Nogo-66 receptor. Science 2002;297 (5584):1190-1193.
    [16]Prinjha R, Moore SE, Vinson M, et al. Inhibitor of neurite outgrowth in humans. Nature 2000;403 (6768):383-384.
    [17]McDonald JW, Sadowsky C. Spinal-cord injury. Lancet 2002;359 (9304):417-425.
    [18]Tator CH, Koyanagi I. Vascular mechanisms in the pathophysiology of human spinal cord injury. J Neurosurg 1997;86 (3):483-492.
    [19]Mautes AE, Weinzierl MR, Donovan F, et al. Vascular events after spinal cord injury:contribution to secondary pathogenesis. Phys Ther 2000;80 (7):673-687.
    [20]Koyanagi I, Tator CH, Lea PJ. Three-dimensional analysis of the vascular system in the rat spinal cord with scanning electron microscopy of vascular corrosion casts. Part 2:Acute spinal cord injury. Neurosurgery 1993;33 (2):285-291; discussion 292.
    [21]Young W. Spinal cord contusion models. Prog Brain Res 2002;137:231-255.
    [22]Doble A. The role of excitotoxicity in neurodegenerative disease:implications for therapy. Pharmacol Ther 1999;81 (3):163-221.
    [23]Choi DW. Ischemia-induced neuronal apoptosis. Curr Opin Neurobiol 1996;6 (5):667-672.
    [24]Park E, Velumian AA, Fehlings MG. The role of excitotoxicity in secondary mechanisms of spinal cord injury:a review with an emphasis on the implications for white matter degeneration. J Neurotrauma 2004;21 (6):754-774.
    [25]Paterniti I, Genovese T, Mazzon E, et al. Liver X receptor agonist treatment regulates inflammatory response after spinal cord trauma. J Neurochem;112 (3):611-624.
    [26]Diaz-Ruiz A, Alcaraz-Zubeldia M, Maldonado V, et al. Differential time-course of the increase of antioxidant thiol-defenses in the acute phase after spinal cord injury in rats. Neurosci Lett 2009;452 (1):56-59.
    [27]Ibarra A, Diaz-Ruiz A. Protective effect of cyclosporin-A in spinal cord injury: an overview. Curr Med Chem 2006; 13 (22):2703-2710.
    [28]Jones TB, McDaniel EE, Popovich PG. Inflammatory-mediated injury and repair in the traumatically injured spinal cord. Curr Pharm Des 2005; 11 (10):1223-1236.
    [29]Popovich PG, Jones TB. Manipulating neuroinflammatory reactions in the injured spinal cord:back to basics. Trends Pharmacol Sci 2003;24 (1):13-17.
    [30]Hulsebosch CE. Recent advances in pathophysiology and treatment of spinal cord injury. Adv Physiol Educ 2002;26 (1-4):238-255.
    [31]Ibarra A, Martinon S. Pharmacological approaches to induce neuroregeneration in spinal cord injury:an overview. Curr Drug Discov Technol 2009;6 (2):82-90.
    [32]Tian DS, Dong Q, Pan DJ, et al. Attenuation of astrogliosis by suppressing of microglial proliferation with the cell cycle inhibitor olomoucine in rat spinal cord injury model. Brain Res 2007; 1154:206-214.
    [33]Tian DS, Xie MJ, Yu ZY, et al. Cell cycle inhibition attenuates microglia induced inflammatory response and alleviates neuronal cell death after spinal cord injury in rats. Brain Res 2007;1135 (1):177-185.
    [34]Xu GY, Hughes MG, Ye Z, et al. Concentrations of glutamate released following spinal cord injury kill oligodendrocytes in the spinal cord. Exp Neurol 2004; 187 (2):329-336.
    [35]Emery E, Aldana P, Bunge MB, et al. Apoptosis after traumatic human spinal cord injury. J Neurosurg 1998;89 (6):911-920.
    [36]Crowe MJ, Bresnahan JC, Shuman SL, et al. Apoptosis and delayed degeneration after spinal cord injury in rats and monkeys. Nat Med 1997;3 (1):73-76.
    [37]McDonald JW, Belegu V. Demyelination and remyelination after spinal cord injury. J Neurotrauma 2006;23 (3-4):345-359.
    [38]Grossman SD, Rosenberg LJ, Wrathall JR. Temporal-spatial pattern of acute neuronal and glial loss after spinal cord contusion. Exp Neurol 2001;168 (2):273-282.
    [39]McTigue DM, Wei P, Stokes BT. Proliferation of NG2-positive cells and altered oligodendrocyte numbers in the contused rat spinal cord. J Neurosci 2001;21 (10):3392-3400.
    [40]Totoiu MO, Keirstead HS. Spinal cord injury is accompanied by chronic progressive demyelination. J Comp Neurol 2005;486 (4):373-383.
    [41]McTigue DM, Tripathi RB. The life, death, and replacement of oligodendrocytes in the adult CNS. J Neurochem 2008;107 (1):1-19.
    [42]Rosenbluth J. A brief history of myelinated nerve fibers:one hundred and fifty years of controversy. J Neurocytol 1999;28 (4-5):251-262.
    [43]Sanchez I, Hassinger L, Paskevich PA, et al. Oligodendroglia regulate the regional expansion of axon caliber and local accumulation of neurofilaments during development independently of myelin formation. J Neurosci 1996; 16 (16):5095-5105.
    [44]Sanchez I, Hassinger L, Sihag RK, et al. Local control of neurofilament accumulation during radial growth of myelinating axons in vivo. Selective role of site-specific phosphorylation. J Cell Biol 2000;151 (5):1013-1024.
    [45]Du Y, Dreyfus CF. Oligodendrocytes as providers of growth factors. J Neurosci Res 2002;68 (6):647-654.
    [46]Wilkins A, Majed H, Layfield R, et al. Oligodendrocytes promote neuronal survival and axonal length by distinct intracellular mechanisms:a novel role for oligodendrocyte-derived glial cell line-derived neurotrophic factor. J Neurosci 2003;23 (12):4967-4974.
    [47]Dai X, Qu P, Dreyfus CF. Neuronal signals regulate neurotrophin expression in oligodendrocytes of the basal forebrain. Glia 2001;34 (3):234-239.
    [48]Wilkins A, Chandran S, Compston A. A role for oligodendrocyte-derived IGF-1 in trophic support of cortical neurons. Glia 2001;36(1):48-57.
    [49]Erschbamer M, Pernold K, Olson L. Inhibiting epidermal growth factor receptor improves structural, locomotor, sensory, and bladder recovery from experimental spinal cord injury. J Neurosci 2007;27 (24):6428-6435.
    [50]Huang X, Kim JM, Kong TH, et al. GM-CSF inhibits glial scar formation and shows long-term protective effect after spinal cord injury. J Neurol Sci 2009;277(1-2):87-97.
    [51]Ferrer I, Alcantara S, Ballabriga J, et al. Transforming growth factor-alpha (TGF-alpha) and epidermal growth factor-receptor (EGF-R) immunoreactivity in normal and pathologic brain. Prog Neurobiol 1996;49 (2):99-123.
    [52]Planas AM, Justicia C, Soriano MA, et al. Epidermal growth factor receptor in proliferating reactive glia following transient focal ischemia in the rat brain. Glia 1998;23 (2):120-129.
    [53]Popovich PG. Immunological regulation of neuronal degeneration and regeneration in the injured spinal cord. Prog Brain Res 2000;128:43-58.
    [54]Bareyre FM, Schwab ME. Inflammation, degeneration and regeneration in the injured spinal cord:insights from DNA microarrays. Trends Neurosci 2003;26 (10):555-563.
    [55]Hisahara S, Shoji S, Okano H, et al. ICE/CED-3 family executes oligodendrocyte apoptosis by tumor necrosis factor. J Neurochem 1997;69 (1):10-20.
    [56]Vartanian T, Li Y, Zhao M, et al. Interferon-gamma-induced oligodendrocyte cell death:implications for the pathogenesis of multiple sclerosis. Mol Med 1995;1 (7):732-743.
    [57]Jurewicz A, Matysiak M, Tybor K, et al. Tumour necrosis factor-induced death of adult human oligodendrocytes is mediated by apoptosis inducing factor. Brain 2005;128 (Pt 11):2675-2688.
    [58]Ladiwala U, Li H, Antel JP, et al. p53 induction by tumor necrosis factor-alpha and involvement of p53 in cell death of human oligodendrocytes. J Neurochem 1999;73 (2):605-611.
    [59]Wosik K, Antel J, Kuhlmann T, et al. Oligodendrocyte injury in multiple sclerosis:a role for p53. J Neurochem 2003;85 (3):635-644.
    [60]Donnelly DJ, Popovich PG. Inflammation and its role in neuroprotection, axonal regeneration and functional recovery after spinal cord injury. Exp Neurol 2008;209 (2):378-388.
    [61]Douglas MR, Morrison KC, Jacques SJ, et al. Off-target effects of epidermal growth factor receptor antagonists mediate retinal ganglion cell disinhibited axon growth. Brain 2009; 132 (Pt 11):3102-3121.
    [62]Albrecht PJ, Murtie JC, Ness JK, et al. Astrocytes produce CNTF during the remyelination phase of viral-induced spinal cord demyelination to stimulate FGF-2 production. Neurobiol Dis 2003;13 (2):89-101.
    [63]Miller RH. Regulation of oligodendrocyte development in the vertebrate CNS. Prog Neurobiol 2002;67 (6):451-467.
    [64]Horner PJ, Power AE, Kempermann G, et al. Proliferation and differentiation of progenitor cells throughout the intact adult rat spinal cord. J Neurosci 2000;20(6):2218-2228.
    [65]Berry M. Post-injury myelin-breakdown products inhibit axonal growth:an hypothesis to explain the failure of axonal regeneration in the mammalian central nervous system. Bibl Anat 1982 (23):1-11.
    [66]Filbin MT. Myelin-associated inhibitors of axonal regeneration in the adult mammalian CNS. Nat Rev Neurosci 2003;4 (9):703-713.
    [67]Fry EJ. Central nervous system regeneration:mission impossible? Clin Exp Pharmacol Physiol 2001;28 (4):253-258.
    [68]GrandPre T, Nakamura F, Vartanian T, et al. Identification of the Nogo inhibitor of axon regeneration as a Reticulon protein. Nature 2000;403 (6768):439-444.
    [69]Benson MD, Romero MI, Lush ME, et al. Ephrin-B3 is a myelin-based inhibitor of neurite outgrowth. Proc Natl Acad Sci U S A 2005; 102 (30):10694-10699.
    [70]Domeniconi M, Cao Z, Spencer T, et al. Myelin-associated glycoprotein interacts with the Nogo66 receptor to inhibit neurite outgrowth. Neuron
    2002;35 (2):283-290.
    [71]Kim JE, Li S, GrandPre T, et al. Axon regeneration in young adult mice lacking Nogo-A/B. Neuron 2003;38 (2):187-199.
    [72]Trapp BD. Myelin-associated glycoprotein. Location and potential functions. Ann N Y Acad Sci 1990;605:29-43.
    [73]Hasegawa Y, Fujitani M, Hata K, et al. Promotion of axon regeneration by myelin-associated glycoprotein and Nogo through divergent signals downstream of Gi/G. J Neurosci 2004;24 (30):6826-6832.
    [74]Huang JK, Phillips GR, Roth AD, et al. Glial membranes at the node of Ranvier prevent neurite outgrowth. Science 2005;310 (5755):1813-1817.
    [75]Vourc'h P, Andres C. Oligodendrocyte myelin glycoprotein (OMgp): evolution, structure and function. Brain Res Brain Res Rev 2004;45 (2):115-124.
    [76]McKerracher L, Winton MJ. Nogo on the go. Neuron 2002;36 (3):345-348.
    [77]王宏,闫慧博,廖华.大剂量甲基强的松龙对大鼠急性损伤脊髓Nogo-A表达量影响的研究.中国临床解剖学杂志,2006;24(3):329-332.
    [1]Rowland JW, Hawryluk GW, Kwon B, et al. Current status of acute spinal cord injury pathophysiology and emerging therapies:promise on the horizon. Neurosurg Focus 2008;25 (5):E2.
    [2]Bareyre FM, Schwab ME. Inflammation, degeneration and regeneration in the injured spinal cord:insights from DNA microarrays. Trends Neurosci 2003;26 (10):555-563.
    [3]Young W. Spinal cord contusion models. Prog Brain Res 2002; 137:231-255.
    [4]Tator CH, Fehlings MG. Review of the secondary injury theory of acute spinal cord trauma with emphasis on vascular mechanisms. J Neurosurg 1991;75 (1):15-26.
    [5]Donnelly DJ, Popovich PG. Inflammation and its role in neuroprotection, axonal regeneration and functional recovery after spinal cord injury. Exp Neurol 2008;209 (2):378-388.
    [6]Sugawara T, Lewen A, Gasche Y, et al. Overexpression of SOD1 protects vulnerable motor neurons after spinal cord injury by attenuating mitochondrial cytochrome c release. Faseb J 2002;16 (14):1997-1999.
    [7]Park E, Velumian AA, Fehlings MG The role of excitotoxicity in secondary mechanisms of spinal cord injury:a review with an emphasis on the implications for white matter degeneration. J Neurotrauma 2004;21 (6):754-774.
    [8]Matute C, Domercq M, Sanchez-Gomez MV. Glutamate-mediated glial injury: mechanisms and clinical importance. Glia 2006;53 (2):212-224.
    [9]Tian DS, Yu ZY, Xie MJ, et al. Suppression of astroglial scar formation and enhanced axonal regeneration associated with functional recovery in a spinal cord injury rat model by the cell cycle inhibitor olomoucine. J Neurosci Res 2006;84(5):1053-1063.
    [10]Li GL, Farooque M, Holtz A, et al. Apoptosis of oligodendrocytes occurs for long distances away from the primary injury after compression trauma to rat spinal cord. Acta Neuropathol 1999;98 (5):473-480.
    [11]Tian DS, Dong Q, Pan DJ, et al. Attenuation of astrogliosis by suppressing of microglial proliferation with the cell cycle inhibitor olomoucine in rat spinal cord injury model. Brain Res 2007; 1154:206-214.
    [12]Pineau I, Lacroix S. Proinflammatory cytokine synthesis in the injured mouse spinal cord:multiphasic expression pattern and identification of the cell types involved. J Comp Neurol 2007;500 (2):267-285.
    [13]Wrathall JR, Teng YD, Choiniere D. Amelioration of functional deficits from spinal cord trauma with systemically administered NBQX, an antagonist of non-N-methyl-D-aspartate receptors. Exp Neurol 1996; 137 (1):119-126.
    [14]Panter SS, Yum SW, Faden AI. Alteration in extracellular amino acids after traumatic spinal cord injury. Ann Neurol 1990;27 (1):96-99.
    [15]Schwartz G, Fehlings MG Evaluation of the neuroprotective effects of sodium channel blockers after spinal cord injury:improved behavioral and neuroanatomical recovery with riluzole. J Neurosurg 2001;94 (2 Suppl):245-256.
    [16]Pearse DD, Chatzipanteli K, Marcillo AE, et al. Comparison of iNOS inhibition by antisense and pharmacological inhibitors after spinal cord injury. J Neuropathol Exp Neurol 2003;62 (11):1096-1107.
    [17]Hains BC, Black JA, Waxman SG Primary cortical motor neurons undergo apoptosis after axotomizing spinal cord injury. J Comp Neurol 2003;462 (3):328-341.
    [18]Bozbuga M, Izgi N, Canbolat A. The effects of chronic alpha-tocopherol administration on lipid peroxidation in an experimental model of acute spinal cord injury. Neurosurg Rev 1998;21 (1):36-42.
    [19]Xiong Y, Rabchevsky AG, Hall ED. Role of peroxynitrite in secondary oxidative damage after spinal cord injury. J Neurochem 2007; 100 (3):639-649.
    [20]Sakamoto A, Ohnishi ST, Ohnishi T, et al. Relationship between free radical production and lipid peroxidation during ischemia-reperfusion injury in the rat brain. Brain Res 1991;554 (1-2):186-192.
    [21]Bao F, Liu D. Peroxynitrite generated in the rat spinal cord induces apoptotic cell death and activates caspase-3. Neuroscience 2003;116 (1):59-70.
    [22]Rosenberg LJ, Teng YD, Wrathall JR. Effects of the sodium channel blocker tetrodotoxin on acute white matter pathology after experimental contusive spinal cord injury. J Neurosci 1999;19 (14):6122-6133.
    [23]Feldblum S, Arnaud S, Simon M, et al. Efficacy of a new neuroprotective agent, gacyclidine, in a model of rat spinal cord injury. J Neurotrauma 2000; 17 (11):1079-1093.
    [24]Fleming JC, Norenberg MD, Ramsay DA, et al. The cellular inflammatory response in human spinal cords after injury. Brain 2006; 129 (Pt 12):3249-3269.
    [25]Bethea JR, Nagashima H, Acosta MC, et al. Systemically administered interleukin-10 reduces tumor necrosis factor-alpha production and significantly improves functional recovery following traumatic spinal cord injury in rats. J Neurotrauma 1999;16 (10):851-863.
    [26]Kim GM, Xu J, Xu J, et al. Tumor necrosis factor receptor deletion reduces nuclear factor-kappaB activation, cellular inhibitor of apoptosis protein 2 expression, and functional recovery after traumatic spinal cord injury. J Neurosci 2001;21 (17):6617-6625.
    [27]Beattie MS, Hermann GE, Rogers RC, et al. Cell death in models of spinal cord injury. Prog Brain Res 2002; 137:37-47.
    [28]Casha S, Yu WR, Fehlings MG Oligodendroglial apoptosis occurs along degenerating axons and is associated with FAS and p75 expression following spinal cord injury in the rat. Neuroscience 2001; 103 (1):203-218.
    [29]Stys PK, Lipton SA. White matter NMDA receptors:an unexpected new therapeutic target? Trends Pharmacol Sci 2007;28 (11):561-566.
    [30]Norenberg MD, Smith J, Marcillo A. The pathology of human spinal cord injury:defining the problems. J Neurotrauma 2004;21 (4):429-440.
    [31]Fitch MT, Silver J. CNS injury, glial scars, and inflammation:Inhibitory extracellular matrices and regeneration failure. Exp Neurol 2008;209 (2):294-301.
    [32]Herrmann JE, Imura T, Song B, et al. STAT3 is a critical regulator of astrogliosis and scar formation after spinal cord injury. J Neurosci 2008;28 (28):7231-7243.
    [33]Hill CE, Beattie MS, Bresnahan JC. Degeneration and sprouting of identified descending supraspinal axons after contusive spinal cord injury in the rat. Exp Neurol 2001;171(1):153-169.
    [34]Kottis V, Thibault P, Mikol D, et al. Oligodendrocyte-myelin glycoprotein (OMgp) is an inhibitor of neurite outgrowth. J Neurochem 2002;82 (6):1566-1569.
    [35]RufF RL, McKerracher L, Selzer ME. Repair and neurorehabilitation strategies for spinal cord injury. Ann N Y Acad Sci 2008; 1142:1-20.
    [36]Prinjha R, Moore SE, Vinson M, et al. Inhibitor of neurite outgrowth in humans. Nature 2000;403 (6768):383-384.
    [37]Grados-Munro EM, Fournier AE. Myelin-associated inhibitors of axon regeneration. J Neurosci Res 2003;74 (4):479-485.
    [38]Coleman MP, Perry VH. Axon pathology in neurological disease:a neglected therapeutic target. Trends Neurosci 2002;25 (10):532-537.
    [39]McMahon SB, Malcangio M. Current challenges in glia-pain biology. Neuron 2009;64 (1):46-54.
    [40]Sievers J, Parwaresch R, Wottge HU. Blood monocytes and spleen macrophages differentiate into microglia-like cells on monolayers of astrocytes:morphology. Glia 1994;12 (4):245-258.
    [41]Nimmerjahn A, Kirchhoff F, Helmchen F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science 2005;308 (5726):1314-1318.
    [42]Lai AY, Todd KG Hypoxia-activated microglial mediators of neuronal survival are differentially regulated by tetracyclines. Glia 2006;53 (8):809-816.
    [43]Beattie MS. Inflammation and apoptosis:linked therapeutic targets in spinal cord injury. Trends Mol Med 2004;10 (12):580-583.
    [44]Block ML, Hong JS. Microglia and inflammation-mediated neurodegeneration: multiple triggers with a common mechanism. Prog Neurobiol 2005;76 (2):77-98.
    [45]Liu J, Fu D, Xu J, et al. [Acute cardiac injury activates interleukin-1 beta signaling in the spinal cord]. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2009;34(3):210-215.
    [46]Sofroniew MV, Vinters HV. Astrocytes:biology and pathology. Acta Neuropathol;119(1):7-35.
    [47]Renault-Mihara F, Okada S, Shibata S, et al. Spinal cord injury:emerging beneficial role of reactive astrocytes' migration. Int J Biochem Cell Biol 2008;40(9):1649-1653.
    [48]Tanaka K, Watase K, Manabe T, et al. Epilepsy and exacerbation of brain injury in mice lacking the glutamate transporter GLT-1. Science 1997;276 (5319):1699-1702.
    [49]Tang X, Davies JE, Davies SJ. Changes in distribution, cell associations, and protein expression levels of NG2, neurocan, phosphacan, brevican, versican V2, and tenascin-C during acute to chronic maturation of spinal cord scar tissue. J Neurosci Res 2003;71 (3):427-444.
    [50]Cafferty WB, Bradbury EJ, Lidierth M, et al. Chondroitinase ABC-mediated plasticity of spinal sensory function. J Neurosci 2008;28 (46):11998-12009.
    [51]McTigue DM, Tripathi RB. The life, death, and replacement of oligodendrocytes in the adult CNS. J Neurochem 2008;107 (1):1-19.
    [52]Rosenbluth J. A brief history of myelinated nerve fibers:one hundred and fifty years of controversy. J Neurocytol 1999;28 (4-5):251-262.
    [53]Wilkins A, Majed H, Layfield R, et al. Oligodendrocytes promote neuronal survival and axonal length by distinct intracellular mechanisms:a novel role for oligodendrocyte-derived glial cell line-derived neurotrophic factor. J Neurosci 2003;23 (12):4967-4974.
    [54]Crowe MJ, Bresnahan JC, Shuman SL, et al. Apoptosis and delayed degeneration after spinal cord injury in rats and monkeys. Nat Med 1997;3 (1):73-76.
    [55]Antel J. Oligodendrocyte/myelin injury and repair as a function of the central nervous system environment. Clin Neurol Neurosurg 2006;108 (3):245-249.
    [56]Jurewicz A, Matysiak M, Tybor K, et al. Tumour necrosis factor-induced death of adult human oligodendrocytes is mediated by apoptosis inducing factor. Brain 2005;128 (Pt 11):2675-2688.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700