LPS腹腔注射诱发大鼠脑内炎症致皮层与海马病理改变及相关机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
包括神经退行性病变在内的多数中枢神经系统疾病均可能与外周炎症过程相关。炎症虽可作为机体抵抗外来刺激的防御性反应,但过度的炎症反应也可造成组织损伤,推动疾病发展。外周炎症过程可导致如发热、厌食等病理反应,提示特定炎症物质可将外周炎症信号传递至中枢神经系统。但这种炎症信号对脑内特定部位,如与认知、学习记忆相关的大脑皮层和海马等区域,是否也有影响,并导致相关的病理改变,其机制如何,至今未见系统报道。
     腹腔注射脂多糖(lipopolysaccharide, LPS)炎症模型中,花生四烯酸代谢产生的炎症物质前列腺素E2(prostaglandin E2, PGE2)和白三烯C4(leukotriene C4, LTC4)作用于下丘脑相关脑区,诱导发热、厌食等症状。由于该类炎症物质可在脑内广泛产生并易于扩散,因而推测,它们也可作用于大脑皮层及海马等与认知、记忆功能密切相关的脑区,并可能作为始动因素诱发下游病理改变。
     然而,在整体炎症模型中,大脑皮层及海马脑区内PGE2和LTC4的生成及其合成酶系表达情况至今未见系统报道,亟需展开相关研究。本研究第一部分采用LPS腹腔注射致脑炎模型,探索致炎后大鼠大脑皮层及海马内该两炎症物质的生成情况,并以环氧合酶抑制剂吲哚美辛(indomethacin, INDO)作为工具药,研究PGE2调控大脑皮层及海马中3-硝基化酪氨酸(3-nitrotyrosine,3-NT)形成,从而论证PGE2作为始动因素推动脑内相关病理改变的作用。
     另一方面,炎症过程诱导生成肿瘤坏死因子α(tumor necrosis factorα, TNF-α), TNF-α参与各种急、慢性中枢神经系统炎症反应。相关研究表明,TNF-α信号通路可上调BACE1表达造成海马脑区内淀粉样蛋白生成增加,进而推动阿尔茨海默病(Alzheimer's disease, AD)的发展。BACE1直接参与淀粉样肽(amyloidb,Aβ)形成,因而是一种具有重要病理意义的标志性蛋白。本研究第二部分采用LPS腹腔注射致脑炎模型及体外培养神经元、星型胶质细胞体系,以TNF-α抑制剂依那西普(etanercept, ETC)为工具药,论证TNF-α信号通路干预BACE1表达的作用及其机制。
     本研究利用体内、外多种炎症生物学体系,系统研究以PGE2、TNF-α为主的炎症物质对大脑内调控情况,既关注以大脑皮层,海马等病理学改变为特征的早期病理学靶点,又关注论证"PGE2-ONOO"-3-NT/BACE1"炎症事件,以期揭示炎症过程推动脑内病理改变的生物学本质,为寻找神经退行性改变相关的药物作用新靶点提供思路。
     1.LPS致大鼠大脑皮质海马PGE2、LTC4合成酶系表达变化及3-硝基酪氨酸形成探索LPS致大鼠脑部炎症模型中,大脑皮层及海马区域PGE2和LTC4合成酶系表达及产物生成情况,并论证其对脑内病理改变的影响。
     LPS (2 mg/kg, i.p.)可致雄性成年SD大鼠体温升高,幅度、时相均与文献报道相符,提示脑部炎症造模成功。大脑皮层、海马组织mPGES-1于24 h内呈时间依赖性表达上调,脑微血管内皮细胞及锥体神经元均可见其表达,PGE2水平同步增高;另一方面,LPS作用1 h后,在基因、蛋白水平,均可见大脑皮层及海马内LTC4S均呈现一过性上调,并伴随LTC4合成能力增强。mGST3表达未受LPS作用影响,未检测出上述脑区内mGST2的表达。
     一氧化氮合酶(nitric oxide synthases, NOSs)及NADPH氧化酶(NADPH oxidases, NOXs)是体内催化NO及ROS的主要酶系。研究发现,LPS致炎1d可导致海马组织内皮型及诱生型NOS (eNOS, iNOS)及神经元特异性NOX4蛋白表达上调,提示海马组织生成NO及ROS。两者反应生成亚硝基阴离子(peroxynitrite, ONOO-),并导致3-NT形成,该过程与AD发病密切相关。免疫组化结果表明,LPS致炎7d可致大鼠海马组织3-NT水平显著上调,提示上述炎症过程可在早期启动或促进AD病程。诱导细胞凋亡是3-NT重要下游毒性机制之一,western blot结果显示,LPS致炎7d伴随海马组织凋亡相关蛋白caspase-8及Bax表达上调。提示3-NT形成可能通过多种途径参与启动细胞凋亡过程。INDO预给药,可抑制LPS引起的动物发热,并部分抑制LPS所致海马组织eNOS、iNOS及NOX4上调,进而缓解海马部位3-NT形成,及caspase-8及Bax上调,提示炎症物质PGE2参与调控NO及ROS生物合成,并经由3-NT诱导的细胞凋亡影响细胞存活。
     细胞丝裂素活化蛋白激酶(mitogen-activated protein kinases, MAPKs)信号通路可在早期参与AD相关的多种分子生物学事件调控。研究发现INDO可在极早期显著抑制LPS诱导的ERK1/2磷酸化激活,但未见其对p38磷酸化水平调控,提示PGE2可能经由激活ERK1/2信号通路参与对iNOS表达调控,而p38信号通路未体现介导PGE2相关的下游分子事件。
     本研究结果表明,外周注射LPS致大鼠脑部炎症模型中,大脑皮层及海马脑区内PGE2及LTC4合成酶系表达发生变化,提示该两炎症物质存在于上述脑区。外周LPS炎症刺激通过诱导PGE2生成,上调海马脑区内NOSs及NOX4表达变化,进而促进3-NT形成,并诱导细胞凋亡倾向增加,提示PGE2作为重要早期因素参与3-NT形成。MAPKs信号通路与上述调控过程有关。
     2.LPS致大鼠炎症经由TNF-α调控星型胶质细胞淀粉样前体蛋白p位剪切酶表达及机制研究
     本研究探索炎症介质TNF-α、PGE2对BACE1表达的调控作用。
     LPS (2 mg/kg, i.p.)可导致大鼠海马组织内BACE1 mRNA、蛋白表达、活性及其酶切产物分泌型淀粉样蛋白β(secreted APPβ, sAPPβ)呈时间依赖性上调。免疫荧光化学复染结果显示,上调BACE1与星型胶质细胞标志蛋白纤维酸性蛋白(glial fibrillary acidic protein, GFAP)高度重合。该结果表明,LPS致大鼠脑部炎症过程中,海马部位星型胶质细胞与上调BACE1密切相关,提示该炎症过程可能经由上调BACE1这一AD病程中的早期关键蛋白推动病程发展,其中星型胶质细胞是介导该作用的主要细胞类型。进一步论研究显示,预给予ETC (5 mg/kg, suc.)可显著降低LPS所致大鼠海马组织BACE1蛋白及sAPPβ表达上调;蛋白定位结果显示,上调的BACE1集中表达在星型胶质细胞中,而不与神经丝表达阳性的神经元共表达。以上结果提示了TNF-α信号通路及星型胶质细胞在LPS所致BACE1上调过程中的重要作用。
     为进一步验证上述结果,将体外培养原代大鼠大脑皮层星型胶质细胞及神经元与TNF-α共孵育。结果表明,TNF-α在10-150 ng/ml范围内,可浓度依赖地上调星型胶质细胞中BACE1蛋白表达,而同浓度TNF-α不影响神经元中BACE1蛋白表达。体内、外实验均提示,TNF-α可通过调控星型胶质细胞中BACE1表达,影响AD病理过程。由于INDO (2 mg/kg, i.p.)未能影响LPS所致大鼠海马BACE1表达上调,提示PGE2并未参与炎症刺激下BACE1表达调控,间接映证了TNF-α对BACE1表达的重要调控作用。
     TNF-α可诱导脑内星型胶质细胞激活、增殖,提示星型胶质细胞中TNF-α信号通路与AD发病密切相关。本研究发现,LPS诱导外周炎症过程中产生的炎症因子TNF-α参与上调星型胶质细胞中BACE1蛋白表达。体外实验结果支持TNF-α信号通路及星型胶质细胞在AD发病过程中的重要作用。
     以上实验表明:
     1.外周注射LPS致大鼠体温升高急性时相内,大脑皮层及海马组织及微血管内皮细胞内mPGES-1表达上调,PGE2水平增加;LTC4S呈现一过性表达升高,未见其同工酶mGST2表达,mGST3表达未受LPS影响。提示腹腔注射LPS可诱导大脑皮层及海马脑区mPGES-1和LTC4S活性增加,产生炎症因子PGE2及LTC4。
     2.外周注射LPS可导致海马组织NOSs、NOX4表达依次上调,NO、ROS水平升高,形成3-NT,并诱发凋亡蛋白表达。INDO可部分抑制上述变化,提示炎症物质PGE2可能作为重要的早期因素参与该病理变化过程。
     3.LPS可致海马内APP剪切酶BACE1表达上调,酶活力和酶切底物sAPPβ均增加,体内外实验均表明TNF-α选择性调控神经组织星型胶质细胞中BACE1表达,提示星型胶质细胞中的TNF-α信号通路在促进AD早期病程中发挥重要作用。
Inflammation process in brain is an important event accompanied with a series of neurodegenerative disorders. The role inflammation played in these disorders has been controversial. Inflammation was usually taken as benefit against extrinsic stimulus, on the contrary, excessive inflammation can also be destructive. Increasing more evidences indicated that inflamation can be a driving force in promoting these neurodegenerative disorders. Pheripheral cytokines or inflammatory factors were unlikely to impact on brain parenchyma due to the existence of blood-brain barrier (BBB). Howerver, prostaglandin E2 (PGE2) and leukotriene C4 (LTC4) derived from arachidonic acid were indicated to get through the BBB and permeated into hypothalamus regions and finally induced pyresis or anorexia. Brains cortex and hippocampus are important regions related with cognition and memory, they are closely related to several neurodegenerative disorders such as Alzheimer's disease (AD). Therefore, it is possible that PGE2 and/or LTC4 may also act on cortex and hippocampus and finally lead to downstream pathological changes in these regions.
     It was remained elusive that the PGE2 and LTC4 production in cortex and hippocampus respectively in a systemic inflammatory model. Therefore, in the first chapter, lipopolysaccharide (LPS) induced inflammation in rats and cycloxygenases (COXs) inhibitor indomethacin (INDO) were employed. The regulation role of PGE2 on early AD-related pathology characterized by 3-nitrotyrosine (3-NT) formation.
     Tumor necrosis factor-α(TNF-α) is an important cytokine produced within brain tissues following inflammatory challenge. TNF-αis involved in acute and chronic inflammation found in neurodegenerative disorders including AD. It was found that TNF-αsignaling impacted onβ-site amyloid precursor protein cleaving enzyme 1 (BACE1) regulation, suggesting TNF-αsignaling may promote AD development. In the second chapter, TNF-αantagonist etanercept (ETC) was employed to investigate the regulation role of TNF-αon BACE1 expression.
     In the present research, several biological systems both in vivo and in vitro were employed to address the potential impact of PGE2 and TNF-αon related pathological events in brain. It was expected to find the mechanisms underlying inflammation impact on brain pathological changes, and further to discover therapeutic targets for neurodegenerative disorders
     1. PGE2 and LTC4 synthases expression changes and 3-nitrotyrosine formation in LPS-challenged rats brain cortex and hippocampus
     To address the potential influence of PGE2 on related pathological changes in brain cortex and hippocampus, rats were challenged by LPS (2 mg/kg, i.p.) injection. Within 24 h injection of LPS, mPGES-1 expression in cortex and hippocampus was up-regulated in a time-dependent manner. LPS-induced fever was inhibited by cycloxygenases inhibitor indomethacin (2 mg/kg, i.p. INDO) injection, suggesting LPS induced PGE2 production in these brain regions, the LPS-challenged inflammatory model has been established. On the other side, LPS challenge induced a transient up-regulation of LTC4S but had no effect on mGST3 expression. Nitric oxide synthases (NOSs) and Nicotinamide-adenine dinucleotide phosphate oxidases (NOXs) are major enzymes catalyze NO and ROS production in vitro, respectively.1 d after LPS challenge, eNOS, iNOS and NOX4 were up-regulated in rats hippocampus, as well as NO and ROS production in tissues. NO and ROS lead to the peroxynitrite (ONOO") production, which further induced 3-nitrotyrosine (3-NT) formation.3-NT formation was suggested to be closed related with AD pathology. Immunohistochemistry analysis revealed that 3-NT level in hippocampus was up-regulated 7 d after LPS challenge, indicating inflammation may progress AD process in an early stage.3-NT is suggested to be involved in cell apoptosis. The apoptosis related proteins caspase-8 and Bax in hippocampus were determined. As results shown, LPS up-regulated caspase-8 and Bax expression 7 d after injection, suggesting 3-NT formation may ultimately induced apoptosis in hippocampus. INDO pre-treatment partly inhibited LPS-caused eNOS, iNOS and NOX4 up-regulation and further reduced 3-NT formation in hippocampus. INDO pre-treatment also partly ameliorated LPS-enhanced caspase-8 and Bax level. These data suggested that PGE2 was involved in NO and ROS production, and further influenced 3-NT formation in hippocampus. These alternations may finally induce apoptosis.
     Mitogen-activated protein kinases, (MAPKs) participate in several AD related molecular events. MAPKs signaling pathways are involved in protein regulations in early stage. The results shown that INDO inhibited LPS-induced ERK1/2 activation, yet the p38 was not affected by INDO intervention. These results suggested that PGE2 may regulated iNOS expression via ERK1/2 but not p38 signaling pathway.
     Overall, current data indicated that peripheral LPS induced PGE2 production in rats hippocampus. PGE2 may up-regulated NOSs and NOX4 expressions, which further lead to 3-NT formation and increased apoptosis tendency, suggesting an initiative role of PGE2 in inflammation induced pathological changes found in several neurodegenerative disorders. MAPKs signaling pathway are involved in these mentioned regulations.
     2. Involvement of TNF-αon BACE1 expression regulations in rats astrocytes, studies in vivo and in vitro.
     Present research was designed to investigate the potential roles of TNF-αand PGE2 on BACE1 expression regulations.
     BACE1 is a key enzyme participate in APP process and ultimately catalyze Aβformation. LPS (2 mg/kg, i.p.) injection led to BACE1 mRNA, protein and enzyme activity up-regulation as well as secreted APP (3 (sAPPβ) production. Double-staining by immunofluorescence indicated the up-regulated BACE1 were highly co-localized with glial fibrillary acidic protein (GFAP) positive astrocytes. These results indicated that LPS-induced inflammation induced astrocytes BACE1 up-regulation in rat hippocampus, suggesting inflammation may promote AD pathology by BACE1 up-regulation. In addition, these results also suggested astrocyte was the principle cell type involved in.
     To identify the role TNF-αplayed in LPS-induced BACE1 up-regulation. ETC was employed. As results shown, pre-treatment with ETC (5 mg/kg, suc.) significantly decreased LPS-induced BACE1 expression and sAPPβproduction in rats hippocampus. Localization trials indicated that BACE1 up-regulation can be observed in GFAP positive astrocytes but not in neurofilament positive neurons. These data indicated that blockage of TNF-αsignaling pathway was effective in reducing LPS-increased BACE1 expression and enzyme activity. To further explore identify the BACE1 regulation by TNF-αsignaling pathway, primary cultured rats astrocytes and neurons were incubated with TNF-αrespectively. As the results shown, after 24 h incubation with TNF-αin a range of 10-150 ng/ml, the BACE1 expression was up-regulated in astrocytes but not in neurons. The data obtained from both in vivo and in vitro suggested that TNF-αmay promote AD pathology by regulating BACE1 expression in astrocytes. TNF-αis a cytokine highly related with AD pathology.
     INDO (2 mg/kg, i.p.) pre-injection had no effects on LPS-induced BACE1 up-regulation, suggesting PGE2 may not involved in BACE1 regulations under inflammatory circumstance.
     Overall, current data collected from both in vivo and in vitro indicated that TNF-αwas involved in astrocytes BACE1 expression regulation, while PGE2 may not be a crucial factor in this process. These data suggested the important roles of TNF-αsignaling pathway and astrocytes in AD pathology.
     Summary
     1. Peripheral injection of LPS induced rats fever instantly, the mPGES-1 in brain cortex and hippocampus was up-rulated in an acute phase, as well as PGE2 content. The LTC4S is the only LTC4 synthase that found can be transiently up-rgulated, indicating that PGE2 may play a more important role in this model as an initial factor.
     2. Peripheral injection of LPS induced NOSs, NOX4 up-regulation, NO, ROS production,3-NT formation and apoptosis related proteins expression in hippocampus in a time sequence. These alternations can be partly inhibited by INDO administration, suggesting that PGE2 was involved in this process.
     3. In LPS-induced braininflammation in rats, TNF-αselectively regulated BACE1 expression in astrocytes both in vivo and in vitro, indicating astrocytes is an important in mediating inflammatory signals to early phase of AD pathology.
引文
1. Cummings, J.L., Alzheimer's disease. N. Engl. J. Med.,2004.351(1):p.56-67.
    2. Engblom, D., et al., Microsomal prostaglandin E synthase-1 is the central switch during immune-induced pyresis. Nat. Neurosci.,2003.6(11):p.1137-8.
    3. Engblom, D., et al., Prostaglandins as inflammatory messengers across the blood-brain barrier. J. Mol. Med.,2002.80(1):p.5-15.
    4. Schroder, O., et al., Microsomal glutathione S-transferases:selective up-regulation of leukotriene C4 synthase during lipopolysaccharide-induced pyresis. Cell. Mol. Life Sci.,2005.62(1):p.87-94.
    5. Luth, H.J., G. Munch and T. Arendt, Aberrant expression of NOS isoforms in Alzheimer's disease is structurally related to nitrotyrosine formation. Brain Res.,2002.953(1-2):p.135-43.
    6. Bagasra, O., et al., Activation of the inducible form of nitric oxide synthase in the brains of patients with multiple sclerosis. Proc. Natl. Acad. Sci. U. S. A.,1995.92(26):p.12041-5.
    7. Martin, L.J., K. Chen and Z. Liu, Adult motor neuron apoptosis is mediated by nitric oxide and Fas death receptor linked by DNA damage and p53 activation. J. Neurosci.,2005.25(27):p.6449-59.
    8. Good, P.F., et al., Protein nitration in Parkinson's disease. J. Neuropathol. Exp. Neurol.,1998. 57(4):p.338-42.
    9. Nunomura, A., et al., Oxidative damage is the earliest event in Alzheimer disease. J. Neuropathol. Exp. Neurol.,2001.60(8):p.759-67.
    10. Aulak, K.S., et al., Proteomic method identifies proteins nitrated in vivo during inflammatory challenge. Proc. Natl. Acad. Sci. U. S. A.,2001.98(21):p.12056-61.
    11. He, P., et al., Deletion of tumor necrosis factor death receptor inhibits amyloid beta generation and prevents learning and memory deficits in Alzheimer's mice. J. Cell Biol.,2007.178(5):p.829-41.
    12. Botchkina, G.I., et al., Expression of TNF and TNF receptors (p55 and p75) in the rat brain after focal cerebral ischemia. Mol. Med.,1997.3(11):p.765-81.
    13. Fillit, H., et al., Elevated circulating tumor necrosis factor levels in Alzheimer's disease. Neurosci. Lett.,1991.129(2):p.318-20.
    14. Butterfield, D.A., et al., Elevated levels of 3-nitrotyrosine in brain from subjects with amnestic mild cognitive impairment:implications for the role of nitration in the progression of Alzheimer's disease. Brain Res.,2007.1148:p.243-8.
    15. Selmaj, K., et al., Tumor necrosis factor-induced proliferation of astrocytes from mature brain is associated with down-regulation of glial fibrillary acidic protein mRNA. J. Neurochem.,1991. 57(3):p.823-30.
    16. Perry, R.T., et al., The role of TNF and its receptors in Alzheimer's disease. Neurobiol. Aging,2001. 22(6):p.873-83.
    17. Smith, M.A., et al., Widespread peroxynitrite-mediated damage in Alzheimer's disease. J. Neurosci., 1997.17(8):p.2653-7.
    18. Wong, M.L., et al., Inducible nitric oxide synthase gene expression in the brain during systemic inflammation. Nat. Med.,1996.2(5):p.581-4.
    19. Vallet, P., et al., Neuronal expression of the NADPH oxidase NOX4, and its regulation in mouse experimental brain ischemia. Neuroscience,2005.132(2):p.233-8.
    20. Blanchard-Fillion, B., et al., Metabolism of 3-nitrotyrosine induces apoptotic death in dopaminergic cells. J. Neurosci.,2006.26(23):p.6124-30.
    21. Ashkenazi, A. and V.M. Dixit, Death receptors:signaling and modulation. Science,1998. 281(5381):p.1305-8.
    22. Chao, D.T. and S.J. Korsmeyer, BCL-2 family:regulators of cell death. Annu. Rev. Immunol.,1998. 16:p.395-419.
    23. Swatton, J.E., et al., Increased MAP kinase activity in Alzheimer's and Down syndrome but not in schizophrenia human brain. Eur. J. Neurosci.,2004.19(10):p.2711-9.
    24. Zhang, J. and S. Rivest, Distribution, regulation and colocalization of the genes encoding the EP2-and EP4-PGE2 receptors in the rat brain and neuronal responses to systemic inflammation. Eur. J. Neurosci.,1999.11(8):p.2651-68.
    25. Chung, K.K. and K.K. David, Emerging roles of nitric oxide in neurodegeneration. Nitric Oxide, 2010.22(4):p.290-5.
    26. Dawson, V.L. and T.M. Dawson, Nitric oxide in neurodegeneration. Prog. Brain Res.,1998.118:p. 215-29.
    27. Paravicini, T.M. and C.G. Sobey, Cerebral vascular effects of reactive oxygen species:recent evidence for a role of NADPH-oxidase. Clin. Exp. Pharmacol. Physiol.,2003.30(11):p.855-9.
    28. Estevez, A.G., et al., Nitric oxide and superoxide contribute to motor neuron apoptosis induced by trophic factor deprivation. J. Neurosci.,1998.18(3):p.923-31.
    29. Mihm, M.J., et al., Free 3-nitrotyrosine causes striatal neurodegeneration in vivo. J. Neurosci., 2001.21(11):p. RC149.
    30. Lancaster, J.R., Jr., Simulation of the diffusion and reaction of endogenously produced nitric oxide. Proc. Natl. Acad. Sci. U. S. A.,1994.91(17):p.8137-41.
    31. Bao, F. and D. Liu, Peroxynitrite generated in the rat spinal cord induces apoptotic cell death and activates caspase-3. Neuroscience,2003.116(1):p.59-70.
    32. Nagata, S., Apoptosis by death factor. Cell,1997.88(3):p.355-65.
    33. Adams, J.M. and S. Cory, The Bcl-2 protein family:arbiters of cell survival. Science,1998. 281(5381):p.1322-6.
    34. Dhillon, A.S., et al., MAP kinase signalling pathways in cancer. Oncogene,2007.26(22):p. 3279-90.
    35. Kim, S.H., J. Kim and R.P. Sharma, Inhibition of p38 and ERK MAP kinases blocks endotoxin-induced nitric oxide production and differentially modulates cytokine expression. Pharmacol. Res.,2004.49(5):p.433-9.
    36. Hu, L.F., et al., Hydrogen sulfide attenuates lipopolysaccharide-induced inflammation by inhibition ofp38 mitogen-activated protein kinase in microglia. J. Neurochem.,2007.100(4):p.1121-8.
    37. Ruano, D., et al., Role of p38 and inducible nitric oxide synthase in the in vivo dopaminergic cells' degeneration induced by inflammatory processes after lipopolysaccharide injection. Neuroscience, 2006.140(4):p.1157-68.
    38. Kim, T.I., et al., The role of mitogen-activated protein kinases and their relationship with NF-kappaB and PPARgamma in indomethacin-Induced apoptosis of colon cancer cells. Ann. N. Y. Acad. Sci.,2002.973:p.241-5.
    39. Wheeler, D., et al., Tumor necrosis factor-alpha-induced neutral sphingomyelinase-2 modulates synaptic plasticity by controlling the membrane insertion of NMDA receptors. J. Neurochem.,2009. 109(5):p.1237-49.
    40. Tobinick, E., Tumour necrosis factor modulation for treatment of Alzheimer's disease:rationale and current evidence. CNS Drugs,2009.23(9):p.713-25.
    41. Deswal, A., et al., Safety and efficacy of a soluble P75 tumor necrosis factor receptor (Enbrel, etanercept) in patients with advanced heart failure. Circulation,1999.99(25):p.3224-6.
    42. Tsimberidou, A.M., et al., Recombinant human soluble tumor necrosis factor (TNF) receptor (p75) fusion protein Enbrel in patients with refractory hematologic malignancies. Cancer Chemother. Pharmacol.,2002.50(3):p.237-42.
    43. Tsimberidou, A.M., et al., Pilot study of recombinant human soluble tumor necrosis factor (TNF) receptor (p75) fusion protein (TNFR:Fc; Enbrel) in patients with refractory multiple myeloma: increase in plasma TNF alpha levels during treatment. Leuk. Res.,2003.27(5):p.375-80.
    44. Rossner, S., et al., Alzheimer's disease beta-secretase BACE1 is not a neuron-specific enzyme. J. Neurochem.,2005.92(2):p.226-34.
    45. Yamamoto, M., et al., Interferon-gamma and tumor necrosis factor-alpha regulate amyloid-beta plaque deposition and beta-secretase expression in Swedish mutant APP transgenic mice. Am. J. Pathol.,2007.170(2):p.680-92.
    46. Hartlage-Rubsamen, M., et al., Astrocytic expression of the Alzheimer's disease beta-secretase (BACE1) is stimulus-dependent. Glia,2003.41(2):p.169-79.
    47. Leuba, G., et al., Neuronal and nonneuronal quantitative BACE immunocytochemical expression in the entorhinohippocampal and frontal regions in Alzheimer's disease. Dement. Geriatr. Cogn. Disord.,2005.19(4):p.171-83.
    48. Griffin, W.S., Perispinal etanercept:potential as an Alzheimer therapeutic. J Neuroinflammation, 2008.5:p.3.
    49. Cutler, R.G., et al., Involvement of oxidative stress-induced abnormalities in ceramide and cholesterol metabolism in brain aging and Alzheimer's disease. Proc. Natl. Acad. Sci. U. S. A., 2004.101(7):p.2070-5.
    50. Jana, A., E.L. Hogan and K. Pahan, Ceramide and neurodegeneration:susceptibility of neurons and oligodendrocytes to cell damage and death. J. Neurol. Sci.,2009.278(1-2):p.5-15.
    51. Adam-Klages, S., et al., Distinct adapter proteins mediate acid versus neutral sphingomyelinase activation through the p55 receptor for tumor necrosis factor. J. Leukoc. Biol.,1998.63(6):p. 678-82.
    52. Eckert, A., et al., Mitochondrial dysfunction, apoptotic cell death, and Alzheimer's disease. Biochem. Pharmacol.,2003.66(8):p.1627-34.
    53. Satoi, H., et al., Astroglial expression of ceramide in Alzheimer's disease brains:a role during neuronal apoptosis. Neuroscience,2005.130(3):p.657-66.
    54. Arboleda, G., et al., Regulation of ceramide-induced neuronal death:cell metabolism meets neurodegeneration. Brain Res Rev,2009.59(2):p.333-46.
    55. Green, D.R. and J.C. Reed, Mitochondria and apoptosis. Science,1998.281(5381):p.1309-12.
    56. Kluck, R.M., et al., The release of cytochrome c from mitochondria:a primary site for Bcl-2 regulation ofapoptosis. Science,1997.275(5303):p.1132-6.
    57. Willaime, S., et al., Ceramide-induced apoptosis in cortical neurons is mediated by an increase in p38 phosphorylation and not by the decrease in ERK phosphorylation. Eur. J. Neurosci.,2001. 13(11):p.2037-46.
    58. Stoica, B.A., et al., Ceramide induces neuronal apoptosis through mitogen-activated protein kinases and causes release of multiple mitochondrial proteins. Mol. Cell. Neurosci.,2005.29(3):p. 355-71.
    59. Kim, S.R., et al., Asiatic acid derivatives enhance cognitive performance partly by improving acetylcholine synthesis. J. Pharm. Pharmacol.,2004.56(10):p.1275-82.
    60. Jeong, B.S., Structure-activity relationship study of asiatic acid derivatives for new wound healing agent. Arch. Pharm. Res.,2006.29(7):p.556-62.
    61. Mook-Jung, I., et al., Protective effects of asiaticoside derivatives against beta-amyloid neurotoxicity. J. Neurosci. Res.,1999.58(3):p.417-25.
    62. Lee, M.K., et al., Asiatic acid derivatives protect cultured cortical neurons from glutamate-induced excitotoxicity. Res. Commun. Mol. Pathol. Pharmacol.,2000.108(1-2):p.75-86.
    63. Krishnamurthy, R.G., et al., Asiatic acid, a pentacyclic triterpene from Centella asiatica, is neuroprotective in a mouse model of focal cerebral ischemia. J. Neurosci. Res.,2009.87(11):p. 2541-50.
    64. Xiong, Y., et al., Protective effects of asiatic acid on rotenone-or H2O2-induced injury in SH-SY5Y cells. Neurochem. Res.,2009.34(4):p.746-54.
    65. Adams, J.D., Jr., Alzheimer's disease, ceramide, visfatin and NAD. CNS Neurol Disord Drug Targets,2008.7(6):p.492-8.
    66. Savioz, A., et al., Contribution of neural networks to Alzheimer disease's progression. Brain Res. Bull.,2009.80(4-5):p.309-14.
    67. Gross, A., et al., Caspase cleaved BID targets mitochondria and is required for cytochrome c release, while BCL-XL prevents this release but not tumor necrosis factor-R1/Fas death. J. Biol. Chem.,1999.274(2):p.1156-63.
    68. Willaime-Morawek, S., et al., IGF-I protects cortical neurons against ceramide-induced apoptosis via activation of the PI-3K/Akt and ERK pathways; is this protection independent of CREB and Bcl-2? Brain Res. Mol. Brain Res.,2005.142(2):p.97-106.
    69. Park, H.J., et al., Beta-amyloid precursor protein is a direct cleavage target of HtrA2 serine protease. Implications for the physiological function of HtrA2 in the mitochondria. J. Biol. Chem., 2006.281(45):p.34277-87.
    70. Benachour, N. and G.E. Seralini, Glyphosate formulations induce apoptosis and necrosis in human umbilical, embryonic, and placental cells. Chem. Res. Toxicol.,2009.22(1):p.97-105.
    71. Yamakura, F., et al., Inactivation of human manganese-superoxide dismutase by peroxynitrite is caused by exclusive nitration of tyrosine 34 to 3-nitrotyrosine. J. Biol. Chem.,1998.273(23):p. 14085-9.
    72. Sanvicens, N. and T.G. Cotter, Ceramide is the key mediator of oxidative stress-induced apoptosis in retinal photoreceptor cells. J. Neurochem.,2006.98(5):p.1432-44.
    1. Hardy, J., The amyloid hypothesis for Alzheimer's disease:a critical reappraisal. J. Neurochem, 2009.110(4):p.1129-34.
    2. Hardy, J. and D. Allsop, Amyloid deposition as the central event in the aetiology of Alzheimer's disease. Trends. Pharmacol. Sci,1991.12(10):p.383-88.
    3. Hardy, J. and D.J. Selkoe, The amyloid hypothesis of Alzheimer's disease:progress and problems on the road to therapeutics. Science,2002.297(5580):p.353-56.
    4. Breitner, J.C., et al., Inverse association of anti-inflammatory treatments and Alzheimer's disease: initial results of a co-twin control study. Neurology,1994.44(2):p.227-32.
    5. Rich, J.B., et al., Nonsteroidal anti-inflammatory drugs in Alzheimer's disease. Neurology,1995. 45(1):p.51-55.
    6. Bonotis, K., et al., Systemic immune aberrations in Alzheimer's disease patients. J Neuroimmunol, 2008.193(1-2):p.183-87.
    7. Fiala, M., et al., Ineffective phagocytosis of amyloid-beta by macrophages of Alzheimer's disease patients. J Alzheimers Dis,2005.7(3):p.221-32; discussion 255-62.
    8. Song,F., et al., Plasma biomarkers for mild cognitive impairment and Alzheimer's disease. Brain Res. Rev,2009.61(2):p.69-80.
    9. Eikelenboom, P., et al., Inflammatory mechanisms in Alzheimer's disease. Trends. Pharmacol. Sci, 1994.15(12):p.447-50.
    10. Mrak, R.E., J.G. Sheng, and W.S. Griffin, Glial cytokines in Alzheimer's disease:review and pathogenic implications. Hum. Pathol,1995.26(8):p.816-23.
    11. Lanzrein, A.S., et al., Longitudinal study of inflammatory factors in serum, cerebrospinal fluid, and brain tissue in Alzheimer disease:interleukin-1 beta, interleukin-6, interleukin-1 receptor antagonist, tumor necrosis factor-alpha, the soluble tumor necrosis factor receptors Ⅰ and Ⅱ, and alpha1-antichymotrypsin. Alzheimer Dis. Assoc. Disord,1998.12(3):p.215-27.
    12. Streit, W.J., et al., Cytokine mRNA profiles in contused spinal cord and axotomized facial nucleus suggest a beneficial role for inflammation and gliosis. Exp. Neurol,1998.152(1):p.74-87.
    13. Cagnin, A., et al., In-vivo measurement of activated microglia in dementia. Lancet,2001.358(9280): p.461-67.
    14. Versijpt, J.J., et al., Assessment of neuroinflammation and microglial activation in Alzheimer's disease with radiolabelled PK11195 and single photon emission computed tomography. A pilot study. Eur. Neurol,2003.50(1):p.39-47.
    15. Licastro, F., et al., Increased plasma levels of interleukin-1, interleukin-6 and alpha-1-antichymotrypsin in patients with Alzheimer's disease:peripheral inflammation or signals from the brain? J. Neuroimmunol,2000.103(1):p.97-102.
    16. Nielsen, H.M., et al., Plasma and CSF serpins in Alzheimer disease and dementia with Lewy bodies. Neurology,2007.69(16):p.1569-79.
    17. Lindberg, C., et al., Soluble interleukin-1 receptor type Ⅱ, IL-18 and caspase-1 in mild cognitive impairment and severe Alzheimer's disease. Neurochem. Int,2005.46(7):p.551-57.
    18. Magaki, S., et al., Increased production of inflammatory cytokines in mild cognitive impairment. Exp. Gerontol,2007.42(3):p.233-40.
    19. Stalder, M., et al., Association of microglia with amyloid plaques in brains of APP23 transgenic mice. Am. J. Pathol,1999.154(6):p.1673-84.
    20. Radde, R., et al., Abeta42-driven cerebral amyloidosis in transgenic mice reveals early and robust pathology. EMBO. Rep,2006.7(9):p.940-46.
    21. Kitazawa, M., et al., Lipopolysaccharide-induced inflammation exacerbates tau pathology by a cyclin-dependent kinase 5-mediated pathway in a transgenic model of Alzheimer's disease. J Neurosci,2005.25(39):p.8843-53.
    22. McGeer, P.L. and E.G. McGeer, Polymorphisms in inflammatory genes and the risk of Alzheimer disease. Arch Neurol,2001.58(11):p.1790-92.
    23. Gallicchio, L., et al., Single nucleotide polymorphisms in inflammation-related genes and mortality in a community-based cohort in Washington County, Maryland. Am. J. Epidemiol,2008.167(7):p. 807-13.
    24. Saunders, A.M., et al., Association of apolipoprotein E allele epsilon 4 with late-onset familial and sporadic Alzheimer's disease. Neurology,1993.43(8):p.1467-72.
    25. Frey, C., et al., Apolipoprotein E epsilon 4 is associated with an increased vulnerability to cell death in Alzheimer's disease. J. Neural. Transm,2006.113(11):p.1753-61.
    26. Levi, O. and D.M. Michaelson, Environmental enrichment stimulates neurogenesis in apolipoprotein E3 and neuronal apoptosis in apolipoprotein E4 transgenic mice. J. Neurochem, 2007.100(1):p.202-10.
    27. Adalbert, R., et al., Abeta, tau and ApoE4 in Alzheimer's disease:the axonal connection. Trends. Mol. Med,2007.13(4):p.135-42.
    28. Kim, J., et al., The role of apolipoprotein E in Alzheimer's disease. Neuron,2009.63(3):p.287-303.
    29. Vitek, M.P., et al., APOE genotype-specific differences in the innate immune response. Neurobio.l Aging,2009.30(9):p.1350-60.
    30. Belinson, H. and D.M. Michaelson, ApoE4-dependent Abeta-mediated neurodegeneration is associated with inflammatory activation in the hippocampus but not the septum. J Neural Transm, 2009.116(11):p.1427-34.
    31. Lipton, S.A., et al., A redox-based mechanism for the neuroprotective and neurodestructive effects of nitric oxide and related nitroso-compounds. Nature,1993.364(6438):p.626-32.
    32. Yamakura, F., et al., Inactivation of human manganese-superoxide dismutase by peroxynitrite is caused by exclusive nitration of tyrosine 34 to 3-nitrotyrosine. J. Biol. Chem,1998.273(23):p. 14085-89.
    33. Nathan, C., et al., Protection from Alzheimer's-like disease in the mouse by genetic ablation of inducible nitric oxide synthase. J. Exp. Med,2005.202(9):p.1163-69.
    34. Smith, M.A., et al., Widespread peroxynitrite-mediated damage in Alzheimer's disease. J. Neurosci, 1997.17(8):p.2653-57.
    35. Sultana, R., et al., Identification of nitrated proteins in Alzheimer's disease brain using a redox proteomics approach. Neurobiol. Dis,2006.22(1):p.76-87.
    36. Nunomura, A., et al., Oxidative damage is the earliest event in Alzheimer disease. J. Neuropathol. Exp. Neurol,2001.60(8):p.759-67.
    37. Butterfield, D.A., Amyloid beta-peptide (1-42)-induced oxidative stress and neurotoxicity: implications for neurodegeneration in Alzheimer's disease brain. A review. Free Radic. Res,2002. 36(12):p.1307-13.
    38. Butterfield, D.A., et al., In vivo oxidative stress in brain of Alzheimer disease transgenic mice: Requirement for methionine 35 in amyloid beta-peptide of APP. Free Radic. Biol. Med,2010.48(1): p.136-44.
    39. Hewett, S.J., et al., Cyclooxygenase-2 contributes to N-methyl-D-aspartate-mediated neuronal cell death in primary cortical cell culture. J. Pharmacol. Exp. Ther,2000.293(2):p.417-25.
    40. Manabe, Y., et al., Prostanoids, not reactive oxygen species, mediate COX-2-dependent neurotoxicity. Ann. Neurol,2004.55(5):p.668-75.
    41. McGeer, P.L. and E.G. McGeer, NSAIDs and Alzheimer disease:epidemiological, animal model and clinical studies. Neurobiol. Aging,2007.28(5):p.639-47.
    42. Szekely, C.A. and P.P. Zandi, Non-Steroidal Anti-Inflammatory Drugs and Alzheimer's Disease:The Epidemiological Evidence. CNS. Neurol. Disord. Drug Targets,2010.
    43. Thirumangalakudi, L., et al., Involvement of PGE2 and PGDH but not COX-2 in thrombin-induced cortical neuron apoptosis. Neurosci. Lett,2009.452(2):p.172-75.
    44. Reed, T.T., et al., Proteomic identification of nitrated brain proteins in early Alzheimer's disease inferior parietal lobule. J Cell Mol Med,2009.13(8B):p.2019-29.
    45. Cimino, P.J., et al., Therapeutic targets in prostaglandin E2 signaling for neurologic disease. Curr. Med. Chem,2008.15(19):p.1863-69.
    46. Hoshino, T., et al., Involvement of prostaglandin E2 in production of amyloid-beta peptides both in vitro and in vivo. J. Biol. Chem,2007.282(45):p.32676-88.
    47. Pooler, A.M., et al., Prostaglandin E2 regulates amyloid precursor protein expression via the EP2 receptor in cultured rat microglia. Neurosci. Lett,2004.362(2):p.127-30.
    48. Kawano, T., et al., Prostaglandin E2 EP1 receptors:downstream effectors of COX-2 neurotoxicity. Nat. Med,2006.12(2):p.225-29.
    49. Engblom, D., et al., Prostaglandins as inflammatory messengers across the blood-brain barrier. J. Mol. Med,2002.80(1):p.5-15.
    50. Carrasco, E., et al., PGE(2) receptor EP1 renders dopaminergic neurons selectively vulnerable to low-level oxidative stress and direct PGE(2) neurotoxicity. J. Neurosci. Res,2007.85(14):p. 3109-17.
    51. Hoshino, T., et al., Prostaglandin E2 stimulates the production of amyloid-beta peptides through internalization of the EP4 receptor. J. Biol. Chem,2009.284(27):p.18493-502.
    52. Tobinick, E., Tumour necrosis factor modulation for treatment of Alzheimer's disease:rationale and current evidence. CNS. Drugs,2009.23(9):p.713-25.
    53. Tarkowski, E., et al., TNF gene polymorphism and its relation to intracerebral production of TNFalpha and TNFbeta in AD. Neurology,2000.54(11):p.2077-81.
    54. McCoy, M.K. and M.G. Tansey, TNF signaling inhibition in the CNS:implications for normal brain function and neurodegenerative disease. J. Neuroinflammation,2008.5:p.45.
    55. Kawano, T., et al., iNOS-derived NO and nox2-derived superoxide confer tolerance to excitotoxic brain injury through peroxynitrite. J. Cereb. Blood Flow. Metab,2007.27(8):p.1453-62.
    56. Luth, H.J., G. Munch, and T. Arendt, Aberrant expression of NOS isoforms in Alzheimer's disease is structurally related to nitrotyrosine formation. Brain Res,2002.953(1-2):p.135-43.
    57. Rogers, J., et al., Clinical trial of indomethacin in Alzheimer's disease. Neurology,1993.43(8):p. 1609-11.
    58. Eriksen, J.L., et al., NSAIDs and enantiomers of flurbiprofen target gamma-secretase and lower Abeta 42 in vivo. J Clin Invest,2003.112(3):p.440-9.
    59. Weggen, S., et al., A subset of NSAIDs lower amyloidogenic Abeta42 independently of cyclooxygenase activity. Nature,2001.414(6860):p.212-16.
    60. Scharf, S., et al., A double-blind, placebo-controlled trial of diclofenac/misoprostol in Alzheimer's disease. Neurology,1999.53(1):p.197-201.
    61. Tobinick, E.L. and H. Gross, Rapid cognitive improvement in Alzheimer's disease following perispinal etanercept administration. J Neuroinflammation,2008.5:p.2

© 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