KA活化的小胶质细胞来源的TNFα对在体和离体海马神经元兴奋毒性作用研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
小胶质细胞(MG)约占胶质细胞总数的20%,属巨噬细胞系,是中枢神经系统病理变化的传感器,在中枢神经系统的损伤,炎症、癫痫等病理过程中均有MG持续活化、增殖现象。MG活化后分泌大量细胞因子和活性分子,包括IL1β, TNFα和NO等,这些过量分泌的物质对神经元具有兴奋毒性损伤作用,与中枢神经系统神经变性疾病(如癫痫)所致神经元的丢失有关。因此活化的MG被普遍认为是一种细胞毒性效应细胞。
     TNFα是一种重要的炎性因子,在脑内神经受损部位,其主要来源于活化的MG。研究表明,在高表达TNFα的转基因小鼠具有年龄依赖性神经变性改变和自发性癫痫;临床使用高剂量的TNFα进行抗肿瘤治疗会诱发癫痫。而使用单克隆抗体拮抗中枢TNFα或抑制小胶质细胞激活,均能有效降低神经元对癫痫的易感性,有助于提高其存活率。
     虽然上述资料提供的证据表明MG活化状态及脑内高水平的TNFα可能与癫痫所致神经元损伤有一定联系,但是,很少有详细的实验资料证明,被活化的MG所分泌的内源性TNFα在海马神经元的兴奋毒性事件中具有重要作用。因此,本研究以致痫剂海人酸(kainic acid, KA)激活MG,以美满霉素(minocycline, MC)作为抑制MG活化的工具药,通过在体和离体实验探讨上述问题。本研究共分三个部分:
     第一部分KA对MG的激活作用及促TNFα分泌效应研究
     为了证实KA对大鼠MG具有激活作用及促进TNFα分泌的效应。本实验先分离纯化培养新生大鼠大脑皮质MG,以100μM KA激活MG或以MC预处理抑制MG的活化,然后应用OX-42免疫染色观察经上述不同处理后MG的形态学变化;分别采用RT-PCR和Western blot法检测MG内TNFαmRNA和蛋白的表达情况;采用Elisα法检测MG条件培养液内TNFα的含量。OX-42免疫组化结果显示,KA作用MG 2hr后,MG胞体增大、突起回缩呈阿米巴状,OX-42的免疫反应较正常组和MC+KA组明显增强。结果显示,MG内TNFαmRNA和蛋白的含量在KA组明显高于正常组;而在MC+KA组则显著低于KA组。Elisα检测结果显示,MG条件培养液中TNFα的含量在KA组明显高于正常组;而MC+KA组则明显低于KA组。结果提示,KA对MG有显著的激活作用,活化的MG合成、分泌TNFα显著增多。MC预处理能抑制MG的活化进而拮抗KA对MG的上述效应。
     第二部分KA活化的MG条件培养液对海马神经元兴奋性的影响及TNFα抗体的拮抗效应
     海马神经元因具有自发放电,兴奋阈值低等特征而与癫痫发病关系密切。中枢神经病变时脑内表达的各种炎性细胞因子能显著影响海马神经元兴奋状态,其机制可能与激活神经元上的相应受体和离子通道有关。在第一部分的实验中,我们已经证实活化的MG合成分泌TNFα显著增多。为了进一步探讨KA活化的MG条件培养液(KA-MCM)对海马神经元的兴奋作用及KA-MCM内的TNFα在这一事件中的重要作用。本实验通过离体和在体实验,采用膜片钳和诱发电位电生理技术、RT-PCR和Western blot方法,研究KA-MCM对海马神经元钙电流、群峰电位、相关兴奋性蛋白NMDAR1、iNOS的影响,并观察TNFα抗体加入KA-MCM后所产生的中和拮抗效应。膜片钳及群峰电位记录表明,KA-MCM能显著增加海马神经元钙电流密度;增强在体大鼠海马CA3区群峰电位。RT-PCR和Western blot检测表明KA-MCM显著增加海马神经元和海马组织NMDAR1和iNOS的mRNA及蛋白质表达,而预先在KA-MCM中加入TNFα抗体则上述效应明显减弱。结果表明,活化的MG来源的TNFα对在体及离体的海马神经元具有极强的神经兴奋作用,由此提示它可能增加机体对癫痫发作及神经元损伤的易感性。
     第三部分KA活化的MG来源的TNFα对海马神经元的促凋亡作用。
     研究表明:在颞叶癫痫患者以及电点燃或化学点燃所致的各种癫痫研究模型中,脑内的病理改变有神经元丧失和与胶质细胞增生,且神经元丧失以神经元凋亡为主要方式。在前二部分实验的基础上,我们进一步探讨KA活化的小胶质细胞培养液(KA-MCM)对海马神经元的促凋亡作用以及TNFα抗体的拮抗效应。本实验通过离体和在体实验,采用原位末端标记(TUNEL)检测,免疫组化、RT-PCR方法,观察KA活化的MG条件培养液(KA-MCM)及TNFα抗体加入KA-MCM后(anti-TNFa-KA-MCM)对海马神经元TUNEL阳性细胞,凋亡关键酶caspase-3 mRNA及其蛋白表达的影响。TUNEL检测结果显示,经UT-MCM、KA-MCM、anti-TNFα-KA-MCM三种不同MCM孵育的海马神经元,在24,48小时时间段其TUNEL阳性细胞数稀少,组间无显著性差别,但在72小时KA-MCM组凋亡细胞显著增多,anti-TNFα-KA-MCM组凋亡细胞数明显少于KA-MCM组。RT-PCR结果显示,大鼠侧脑室注射KA-MCM48小时海马caspase-3 mRNA表达有所增加,72小时明显增加,相同时间点anti-TNFa-KA-MCM组caspase-3 mRNA表达较KA-MCM组明显减弱。caspase-3免疫组化结果与上述RT-PCR结果趋势相似。结果表明,活化的MG来源的TNFα对在体及离体海马神经元具有明显的促凋亡作用,可通过激活caspase-3参与介导该过程。
     小结:KA活化的MG高表达和分泌TNFαKA-MCM可显著增强大鼠在体海马群峰电位;增加离体海马神经元钙电流密度以及海马神经元或海马组织NMDAR1,iNOS和caspase-3 mRNA和蛋白表达;经KA-MCM孵育的海马神经元凋亡细胞显著增多。而若在KA-MCM中预先加入抗TNFα抗体,可明显抑制上述效应。本研究提供的上述结果表明,KA活化的MG来源的TNF-α在离体和在体的情况下,具有神经兴奋毒性作用,可诱导海马神经元凋亡发生。根据本实验结果,我们认为TNFα对海马神经元的兴奋毒性作用可能与激活神经元谷氨酸受体及电压门控钙通道,使胞内钙超载,一氧化氮(NO)过度生成、激活凋亡关键酶caspase-3等多种因素有关。神经免疫调节机制在癫痫的治疗研究领域是近年备受关注的热点,本研究为临床癫痫防治、合理开发神经保护药物提供了新的实验资料和理论依据。
Microglia constitute 20% of the total glial cell population and belong to macrophages cell line. They are sensors for pathological events in the CNS. Sustained activation and proliferation of microglia were observed during the pathological progressions of injuries, inflammation and epilepsy in CNS.Activated microglia secret a variety of proinflammatory cytokine and toxic molecular, include IL1β, TNFa and NO et al. Those over secreted substance have excito-toxic effects on neurons and which are associated with neurpnal loss in neurodegeneration disease such as epilepsy. So activated MG are generally recognized as cytotoxic effecter cells.
     TNFa is an important inflammatory mediator which is mainly derived from MG at the site of injured neurons in the brain. Over expression of brain TNFa in transgenic mice is associated with the occurrence of age-dependent neurodegenerative changes and sporadic spontaneous seizures. It have been found that High dose of TNFa during anti-tumor therapy in clinic could induce seizures in human. However, antagonisms of TNFa using a monoclonal antibody or inhibition of MG activation decreases the seizure susceptibility of neurons and improves their survival rate..
     Although the evidence provided by above materials indicates that the activation state of MG and the high level of TNFa in the brain may have relationship with the seizure-induced neuronal injury, fewer detailed studies have demonstrated that the TNFa derived from activated microglia may play an important role in the event of excitotoxicity of hippocampal neurons. Therefore, in present study, KA was used as an activator and minocycline was used as a tool drug to inhibit the activation of microglia. This study is divided into three parts to explore the above matter through in vivo and in vitro experiments.
     To confirm that KA can activate rat's MG and promote its secretion, MG from neonatal rat cerebral cortex were separated, purified and cultured.100μM KA was used to activate MG and minocycline pretreatment was used to inhibit the activation of MG. Then OX-42 immunostaining was performed to observing the morphological changes of MG undergoing above different treatments. RT-PCR and western blot was applied respectively to detecting the expression of TNFa mRNA and protein; Elisa was performed to testing the content of TNFa in microglial conditioned medium. The results of OX-42 immunohistochemistry showed that after being treated with KA for 2 hours, MG showed amoeba like shape with expanded cell bodies and retracted neurites, it's OX-42 immunoreaction was obviously enhanced than those of the control group and the MC+KA group. The results of RT-PCR and western blot showed that the content of TNFa mRNA and protein in MG in KA group were obviously higher than those of the control group, and which was lower in MC+KA group than in KA group. The result of Elisa showed that TNFa levels in microglial conditioned media of KA group was significantly higher than those of the control group, whereas it was lower in MC+KA group than in KA group. The results indicate that KA significantly activated MG, and TNFa is over synthesized and secreted by activated MG. Pre-treatment with minocycline can inhibit MG activation and further antagonize above effects of KA.
     Because of the features of auto-discharge and low excitation threshold, hippocampal neurons have a close relationship with pathogenesis of epilepsy. A variety of cytokines expressed in the brain during the process of CNS diseases have an obvious effect on the excitability of hippocampal neurons, which may be rely on a mechanism of activation of their respective receptors and ion channels on neurons. In the first part of the study, we have confirmed that the synthesis and secretion of TNFa are increased in activated MG. To further explore the excitatory effects of KA-MCM on hippocampal neurons as well as the important role that KA-MCM contained TNFa plays in this event. Through in vitro and in vivo studies, we used patch clamp and evoked potential electrophysiological technology、RT-PCR and Western blot methods to study the effects of KA-MCM on hippocampal neuronal calcium current, population spike and the expression of related excitatory protein (NMDAR1 and iNOS). The antagonisms effects of TNFa antibody was observed when it was added into KA-MCM. The records of Patch clamp and population spike showed that KA-MCM significantly increased calcium current density in hippocampal neurons, and enhanced population spike on hippocampal CA3 region of rats in vivo. RT-PCR and Western blot showed that KA-MCM significantly enhanced the expression of mRNA and protein of NMDAR1 and iNOS in hippocampal neurons and tissues, whereas addition of anti-TNFa to KA-MCM significantly eliminated such effects. The results indicated that activated MG derived TNFa have strong excitatory effects on hippocampal neurons in vivo and in vitro and which can increase the body susceptibility to seizure and neuronal injury.
     Studies showed that pathological changes of brain include neuron loss and gliocyte hyperplasy in chemical or electrical kindling model of epilepsy as well as patients with temporal lobe epilepsy, and apoptosis is the major way of neuronal loss. Based on above two part of the studies, here we further explore the pro-apoptosis effects of KA-MCM in hippocampal neurons and the antagonistic effects of anti-TNFa. Through in vitro and in vivo studies, by using TUNEL detection, immunohistochemistry and RT-PCR method, we observe the effects of KA-MCM and anti-TNFα-KA-MCM (adding anti-TNFa into KA-MCM) on the expressions of TUNEL positive cells as well as caspase-3 mRNA and protein. The result of TUNEL detection showed that there are few TUNEL positive cells and no significant difference among them at 24,48 hr time period after incubated with three different kinds of MCMs, but they are obviously increased at 72 hr after incubated with KA-MCM. Apoptotic cells in anti-TNFa-KA-MCM group are less than those of KA-MCM group. The result of RT-PCR showed that the expression of caspase-3 mRNA is increased at 48 hr and increased obviously in 72 hr after intra-cerebroventricular injection of KA-MCM. At same time point they are less expressed in anti-TNFa-KA-MCM than in KA-MCM group. The result of caspase-3 immunohistochemistry has the same tendency with above result of RT-PCR. The results indicate that the TNFa derived from KA activated microglia can significantly promote apoptosis of hippocampal neurons. The activation of the caspase-3 is involved in this process.
     Summary:KA activated MG highly expressed and secreted TNFa. KA-MCM can significantly enhance hippocampal population spike in vivo, and increase calcium current、the expression of mRNA and protein of NMDAR1, iNOS、and caspase-3 of hippocampal neurons or hippocampal tissue in vitro. Apoptosis cells are increased significantly after being incubated with KA-MCM. However, the above effects of KA-MCM can be inhibited by adding anti-TNFa antibody into KA-MCM. The above results provided by present research indicate that TNFa derived from KA activated microglia is neuroexcitoxic to hippocampal neurons in vitro and in vivo, which can induce apoptosis on hippocampal neurons. On the basis of the results, We consider that the excito-toxic effect of TNFa on hippocampal neuron may be related to the activation of Glu receptors and voltage-gated calcium channels which result in calcium overload, nitric oxide (NO) production, and activation of caspase-3 (a key enzyme of apoptotic death), el al. The mechanism of neural-immune regulation is a hot spot of concern in therapy and study areas in recently years. This study provides new experimental materials and theory supports for the prevention and therapy of epilepsy, as well as the rational design of neuroprotective drugs.
引文
1. Vezzani, A., S. Balosso, and T. Ravizza, The role of cytokines in the pathophysiology of epilepsy. Brain Behav Immun,2008.22(6):p.797-803.
    2. Mittelman, A., et al., A phase I pharmacokinetic study of recombinant human tumor necrosis factor administered by a 5-day continuous infusion. Invest New Drugs,1992. 10(3):p.183-90.
    3. McCarthy, K.D. and J. de Vellis, Preparation of separate astroglial and oligodendroglial cell cultures from rat cerebral tissue. J Cell Biol,1980.85(3):p. 890-902.
    4. Tikka, T., et al., Minocycline, a tetracycline derivative, is neuroprotective against excitotoxicity by inhibiting activation and proliferation of microglia. J Neurosci,2001. 21(8):p.2580-8.
    5. Elkabes, S., E.M. DiCicco-Bloom, and I.B. Black, Brain microglia/macrophages express neurotrophins that selectively regulate microglial proliferation and function. J Neurosci,1996.16(8):p.2508-21.
    6. Somera-Molina, K.C., et al., Glial activation links early-life seizures and long-term neurologic dysfunction:evidence using a small molecule inhibitor of proinflammatory cytokine upregulation. Epilepsia,2007.48(9):p.1785-800.
    7. Suk, K., Minocycline suppresses hypoxic activation of rodent microglia in culture. Neurosci Lett,2004.366(2):p.167-71.
    8. Giuliani, F., W. Hader, and V.W. Yong, Minocycline attenuates T cell and microglia activity to impair cytokine production in T cell-microglia interaction. J Leukoc Biol, 2005.78(1):p.135-43.
    9. Filipovic, R. and N. Zecevic, Neuroprotective role of minocycline in co-cultures of human fetal neurons and microglia. Exp Neurol,2008.211(1):p.41-51.
    10. Noda, M., et al., AMPA-kainate subtypes of glutamate receptor in rat cerebral microglia. J Neurosci,2000.20(1):p.251-8.
    11. Rappold, P.M., E. Lynd-Balta, and S.A. Joseph, P2X7 receptor immunoreactive profile confined to resting and activated microglia in the epileptic brain. Brain Res, 2006.1089(1):p.171-8.
    12. Shandra, A. A., et al., The role of TNF-alpha in amygdala kindled rats. Neurosci Res, 2002.42(2):p.147-53.
    1. Turrin, N.P. and S. Rivest, Innate immune reaction in response to seizures: implications for the neuropathology associated with epilepsy. Neurobiol Dis, 2004.16(2):p.321-34.
    2. Schafers, M. and L. Sorkin, Effect of cytokines on neuronal excitability. Neurosci Lett,2008.437(3):p.188-93.
    3. Yang, S., et al., Effects of Liuwei Dihuang decoction on ion channels and synaptic transmission in cultured hippocampal neuron of rat. J Ethnopharmacol, 2006.106(2):p.166-72.
    4. Moulder, K.L., et al., Ethanol-induced death of postnatal hippocampal neurons. Neurobiol Dis,2002.10(3):p.396-409.
    5. Palizvan, M.R., Y. Fathollahi, and S. Semnanian, Epileptogenic insult causes a shift in the form of long-term potentiation expression. Neuroscience,2005. 134(2):p.415-23.
    6. DeLorenzo, R.J., S. Pal, and S. Sombati, Prolonged activation of the N-methyl-D-aspartate receptor-Ca2+ transduction pathway causes spontaneous recurrent epileptiform discharges in hippocampal neurons in culture. Proc Natl Acad Sci U S A,1998.95(24):p.14482-7.
    7. Dolphin, A.C., A short history of voltage-gated calcium channels. Br J Pharmacol, 2006.147 Suppl 1:p. S56-62.
    8. Pickering, M., D. Cumiskey, and J.J. O' Connor, Actions of TNF-alpha on glutamatergic synaptic transmission in the central nervous system. Exp Physiol, 2005.90(5):p.663-70.
    9. Czeschik, J.C., et al., TNF-alpha differentially modulates ion channels of nociceptive neurons. Neurosci Lett,2008.434(3):p.293-8.
    10. Stellwagen, D., et al., Differential regulation of AMPA receptor and GABA receptor trafficking by tumor necrosis factor-alpha. J Neurosci,2005.25(12):p. 3219-28.
    11. Ogoshi, F., et al., Tumor necrosis-factor-alpha (TNF-alpha) induces rapid insertion of Ca2+-permeable alpha-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA)/kainate (Ca-A/K) channels in a subset of hippocampal pyramidal neurons. Exp Neurol,2005.193(2):p.384-93.
    12. Leonoudakis, D., et al., TNFalpha-induced AMPA-receptor trafficking in CNS neurons; relevance to excitotoxicity? Neuron Glia Biol,2004.1(3):p.263-273.
    13. Vezzani, A., S. Balosso, and T. Ravizza, The role of cytokines in the pathophysiology of epilepsy. Brain Behav Immun,2008.22(6):p.797-803.
    14. Norris, C.M., et al., Electrophysiological mechanisms of delayed excitotoxicity: positive feedback loop between NMD A receptor.current and depolarization-mediated glutamate release. J Neurophysiol,2006.96(5):p.2488-500.
    15. Deshpande, L.S., et al., Time course and mechanism of hippocampal neuronal death in an in vitro model of status epilepticus:role of NMDA receptor activation and NMDA dependent calcium entry. Eur J Pharmacol,2008.583(1):p.73-83.
    16. Dawson, V.L., et al., Nitric oxide mediates glutamate neurotoxicity in primary cortical cultures. Proc Natl Acad Sci U S A,1991.88(14):p.6368-71.
    17. Lee, S.M., et al., Minocycline inhibits apoptotic cell death via attenuation of TNF-alpha expression following iNOS/NO induction by lipopolysaccharide in neuron/glia co-cultures. J Neurochem,2004.91(3):p.568-78.
    1. Sugama, S., et al., Possible roles of microglial cells for neurotoxicity in clinical neurodegenerative diseases and experimental animal models. Inflamm Allergy Drug Targets,2009.8(4):p.277-84.
    2. Takeuchi, H., et al., Neuritic beading induced by activated microglia is an early feature of neuronal dysfunction toward neuronal death by inhibition of mitochondrial respiration and axonal transport. J Biol Chem,2005.280(11):p.10444-54.
    3. Nagatsu, T. and M. Sawada, Cellular and molecular mechanisms of Parkinson's disease:neurotoxins, causative genes, and inflammatory cytokines. Cell Mol Neurobiol,2006.26(4-6):p.781-802.
    4. Von Bernhardi, R. and J. Eugenin, Microglial reactivity to beta-amyloid is modulated by astrocytes and proinflammatory factors. Brain Res,2004.1025(1-2):p.186-93.
    5. Venters, H.D., R. Dantzer, and K.W. Kelley, A new concept in neurodegeneration: TNFalpha is a silencer of survival signals. Trends Neurosci,2000.23(4):p.175-80.
    6. Chao, C.C. and S. Hu, Tumor necrosis factor-alpha potentiates glutamate neurotoxicity in human fetal brain cell cultures. Dev Neurosci,1994.16(3-4):p. 172-9.
    7. Henshall, D.C., Apoptosis signalling pathways in seizure-induced neuronal death and epilepsy. Biochem Soc Trans,2007.35(Pt 2):p.421-3.
    8. Henshall, D.C. and R.P. Simon, Epilepsy and apoptosis pathways. J Cereb Blood Flow Metab,2005.25(12):p.1557-72.
    9. Xu, S., et al., Neuronal apoptosis in the resected sclerotic hippocampus in patients with mesial temporal lobe epilepsy. J Clin Neurosci,2007.14(9):p.835-40.
    10. Choi, J., et al., Cellular injury and neuroinflammation in children with chronic intractable epilepsy. J Neuroinflammation,2009.6:p.38.
    11. Penkowa, M., et al., Interleukin-6 deficiency reduces the brain inflammatory response and increases oxidative stress and neurodegeneration after kainic acid-induced seizures. Neuroscience,2001.102(4):p.805-18.
    12. Gawlowicz, M., et al., Apoptotic markers in various stages of amygdala kindled seizures in rats. Pharmacol Rep,2006.58(4):p.512-8.
    13. Gastard, M.C., J.C. Troncoso, and V.E. Koliatsos, Caspase activation in the limbic cortex of subjects with early Alzheimer's disease. Ann Neurol,2003.54(3):p.393-8.
    14. Kondratyev, A. and K. Gale, Intracerebral injection of caspase-3 inhibitor prevents neuronal apoptosis after kainic acid-evoked status epilepticus. Brain Res Mol Brain Res,2000.75(2):p.216-24.
    15. Narkilahti, S., et al., Expression and activation of caspase 3 following status epilepticus in the rat. Eur J Neurosci,2003.18(6):p.1486-96.
    1. Benveniste, E.N., Role of macrophages/microglia in multiple sclerosis and experimental allergic encephalomyelitis. J Mol Med,1997.75(3):p.165-73.
    2. Van Furth, R., Phagocytic cells:Development and distribution of mononuclear phagocytes in normal steady state and inflammation. In Inflammation:Basic Principles and Clinical Correlates, ed. G.I. Gallin JI, Snydermann R.1988, New York: Raven Press.281-295.
    3. Brockhaus, J., T. Moller, and H. Kettenmann, Phagocytozing ameboid microglial cells studied in a mouse corpus callosum slice preparation. Glia,1996.16(1):p.81-90.
    4. Polazzi, E. and A. Contestabile, Reciprocal interactions between microglia and neurons:from survival to neuropathology. Rev Neurosci,2002.13(3):p.221-42.
    5. Fagan, A.M. and F.H. Gage, Cholinergic sprouting in the hippocampus:a proposed role for IL-1. Exp Neurol,1990.110(1):p.105-20.
    6. Beattie, E.C., et al., Control of synaptic strength by glial TNFalpha. Science,2002. 295(5563):p.2282-5.
    7. Stalder, A.K., et al., Late-onset chronic inflammatory encephalopathy in immune-competent and severe combined immune-deficient (SCID) mice with astrocyte-targeted expression of tumor necrosis factor. Am J Pathol,1998.153(3):p. 767-83.
    8. Zheng, H., et al., Kainic acid-activated microglia mediate increased excitability of rat hippocampal neurons in vitro and in vivo:crucial role of interleukin-1beta. Neuroimmunomodulation.17(1):p.31-8.
    9. Brown, GC. and A. Bal-Price, Inflammatory neurodegeneration mediated by nitric oxide, glutamate, and mitochondria. Mol Neurobiol,2003.27(3):p.325-55.
    10. Sharma, G. and S. Vijayaraghavan, Nicotinic cholinergic signaling in hippocampal astrocytes involves calcium-induced calcium release from intracellular stores. Proc Natl Acad Sci U S A,2001.98(7):p.4148-53.
    11. Voutsinos-Porche, B., et al., Glial glutamate transporters mediate a functional metabolic crosstalk between neurons and astrocytes in the mouse developing cortex. Neuron,2003.37(2):p.275-86.
    12. Gomes, F.C., D. Paulin, and V. Moura Neto, Glial fibrillary acidic protein (GFAP): modulation by growth factors and its implication in astrocyte differentiation. Braz J Med Biol Res,1999.32(5):p.619-31.
    13. Trejo, F., et al., Gene therapy in a rodent model of Parkinson's disease using differentiated C6 cells expressing a GFAP-tyrosine hydroxylase transgene. Life Sci, 1999.65(5):p.483-91.
    14. Albrecht, P.J., et al., Ciliary neurotrophic factor activates spinal cord astrocytes, stimulating their production and release of fibroblast growth factor-2, to increase motor neuron survival. Exp Neurol,2002.173(1):p.46-62.
    15. Emsley, J.G., P. Arlotta, and J.D. Macklis, Star-cross'd neurons:astroglial effects on neural repair in the adult mammalian CNS. Trends Neurosci,2004.27(5):p.238-40.
    16. Taga, T. and S. Fukuda, Role of IL-6 in the neural stem cell differentiation. Clin Rev Allergy Immunol,2005.28(3):p.249-56.
    17. Weigao, S., Research Progress of Biology Function of Astrocyte and Its Relationship with Disease. Journal of Beihua University(Natural Science),2008.9(6):p.501-509.
    18. Pavelko, K.D., et al., Interleukin-6 protects anterior horn neurons from lethal virus-induced injury. J Neurosci,2003.23(2):p.481-92.
    19. Gabriel, C., et al., Transforming growth factor alpha-induced expression of type 1 plasminogen activator inhibitor in astrocytes rescues neurons from excitotoxicity. FASEB J,2003.17(2):p.277-9.
    20. Min, K.J., et al., Protein kinase A mediates microglial activation induced by plasminogen and gangliosides. Exp Mol Med,2004.36(5):p.461-7.
    21. Dong, Y. and E.N. Benveniste, Immune function of astrocytes. Glia,2001.36(2):p. 180-90.
    22. Kai, W., Astrocyte and nerve functional recovery. Chinese Journal Of Clinical Rehabilitation 2006.10(4):p.158-159.
    23. Ullian, E.M., et al., Control of synapse number by glia. Science,2001.291(5504):p. 657-61.
    24. Bacci, A., et al., The role of glial cells in synaptic function. Philos Trans R Soc Lond B Biol Sci,1999.354(1381):p.403-9.
    25. Song, H., C.F. Stevens, and F.H. Gage, Astroglia induce neurogenesis from adult neural stem cells. Nature,2002.417(6884):p.39-44.
    26.王国卿,杜景卫,夏作理,星形胶质细胞生物学功能和活化.泰山医学院学报,2005.26(2):p.184-186.
    27. Deng, W. and R.D. Poretz, Oligodendroglia in developmental neurotoxicity. Neurotoxicology,2003.24(2):p.161-78.
    28. Sortwell, C.E., et al., Oligodendrocyte-type 2 astrocyte-derived trophic factors increase survival of developing dopamine neurons through the inhibition of apoptotic cell death. J Comp Neurol,2000.426(1):p.143-53.
    29. Du, Y. and C.F. Dreyfus, Oligodendrocytes as providers of growth factors. J Neurosci Res,2002.68(6):p.647-54.
    30. McKerracher, L. and M.J. Winton, Nogo on the go. Neuron,2002.36(3):p.345-8.
    31. Levison, S. W., et al., Hypoxia/ischemia depletes the rat perinatal subventricular zone of oligodendrocyte progenitors and neural stem cells. Dev Neurosci,2001.23(3):p. 234-47.
    32.朱长庚,胶质细胞与神经元间的信号交流及其与癫痫发病机制的关系.中国组织化学与细胞化学杂志,2003.12(3):p.323-326.
    33. Zhu, Y, et al., Transforming growth factor-beta 1 increases bad phosphorylation and protects neurons against damage. J Neurosci,2002.22(10):p.3898-909.
    34. Takeuchi, H., et al., Neuritic beading induced by activated microglia is an early feature of neuronal dysfunction toward neuronal death by inhibition of mitochondrial respiration and axonal transport. J Biol Chem,2005.280(11):p.10444-54.
    35. Rogers, J., et al., Complement activation by beta-amyloid in Alzheimer disease. Proc Natl Acad Sci U S A,1992.89(21):p.10016-20.
    36. Matsuoka, Y, et al., Inflammatory responses to amyloidosis in a transgenic mouse model of Alzheimer's disease. Am J Pathol,2001.158(4):p.1345-54.
    37. Strohmeyer, R., Y Shen, and J. Rogers, Detection of complement alternative pathway mRNA and proteins in the Alzheimer's disease brain. Brain Res Mol Brain Res,2000. 81(1-2):p.7-18.
    38. Davis, E.J., T.D. Foster, and W.E. Thomas, Cellular forms and functions of brain microglia. Brain Res Bull,1994.34(1):p.73-8.
    39. Kim, S.U. and J. de Vellis, Microglia in health and disease. J Neurosci Res,2005. 81(3):p.302-13.
    40. Walker, D.G. and L.F. Lue, Investigations with cultured human microglia on pathogenic mechanisms of Alzheimer's disease and other neurodegenerative diseases. J Neurosci Res,2005.81(3):p.412-25.
    41. Blume, A.J., Vitek, M. P, Focusing on IL-1-promotion of β-amyloid precursor protein synthesis as an early event in Alzheimer's disease. Neurobiol. Aging,1989.10:p. 406-408.
    42. Weiner, H.L. and D. Frenkel, Immunology and immunotherapy of Alzheimer's disease. Nat Rev Immunol,2006.6(5):p.404-16.
    43. Mirza, B., et al., The absence of reactive astrocytosis is indicative of a unique inflammatory process in Parkinson's disease. Neuroscience,2000.95(2):p.425-32.
    44. Ouchi, Y, et al., Microglial activation and dopamine terminal loss in early Parkinson's disease. Ann Neurol,2005.57(2):p.168-75.
    45. He, Y, S. Appel, and W. Le, Minocycline inhibits microglial activation and protects nigral cells after 6-hydroxydopamine injection into mouse striatum. Brain Res,2001. 909(1-2):p.187-93.
    46. Du, Y., et al., Minocycline prevents nigrostriatal dopaminergic neurodegeneration in the MPTP model of Parkinson's disease. Proc Natl Acad Sci U S A,2001.98(25):p. 14669-74.
    47. Lee, J.C., et al., Intracerebral hemorrhage-induced brain injury is aggravated in senescence-accelerated prone mice. Stroke,2006.37(1):p.216-22.
    48. Kowianski, P., et al., Evolution of microglial and astroglial response during experimental intracerebral haemorrhage in the rat. Folia Neuropathol,2003.41(3):p. 123-30.
    49. Ross, F.M., et al., Carbenoxolone depresses spontaneous epileptiform activity in the CA1 region of rat hippocampal slices. Neuroscience,2000.100(4):p.789-96.
    50. Kozlov, A.S., et al., Target cell-specific modulation of neuronal activity by astrocytes. Proc Natl Acad Sci U S A,2006.103(26):p.10058-63.
    51. Kimura, N., et al., Amyloid beta up-regulates brain-derived neurotrophic factor production from astrocytes:rescue from amyloid beta-related neuritic degeneration. J Neurosci Res,2006.84(4):p.782-9.
    52. Sacha, P., et al., Expression of glutamate carboxypeptidase Ⅱ in human brain. Neuroscience,2007.144(4):p.1361-72.
    53. Ralay Ranaivo, H., et al., Glia as a therapeutic target:selective suppression of human amyloid-beta-induced upregulation of brain proinflammatory cytokine production attenuates neurodegeneration. J Neurosci,2006.26(2):p.662-70.
    54. Lucchinetti, C., et al., A quantitative analysis of oligodendrocytes in multiple sclerosis lesions. A study of 113 cases. Brain,1999.122 (Pt 12):p.2279-95.
    55. Kurosinski, P. and J. Gotz, Glial cells under physiologic and pathologic conditions. Arch Neurol,2002.59(10):p.1524-8.
    56. Wolswijk, G, Oligodendrocyte precursor cells in the demyelinated multiple sclerosis spinal cord. Brain,2002.125(Pt 2):p.338-49.
    57. Masumura, M., et al., Oligodendroglial cell death with DNA fragmentation in the white matter under chronic cerebral hypoperfusion:comparison between normotensive and spontaneously hypertensive rats. Neurosci Res,2001.39(4):p. 401-12.
    58. Mabuchi, T., et al., Contribution of microglia/macrophages to expansion of infarction and response of oligodendrocytes after focal cerebral ischemia in rats. Stroke,2000. 31(7):p.1735-43.

© 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