鱼藤酮对PC12细胞毒性作用机制的蛋白质组学研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
本研究从蛋白质组学的角度探讨帕金森病(Parkinson’s disease,PD)的可能病理机制,筛选PD的关键蛋白,期望发现PD的分子标志,为神经防护药物治疗提供靶点。本研究采用MTT法和荧光法检测鱼藤酮诱导的PC12细胞PD模型,首次应用差异凝胶电泳(DIGE)和质谱(MS)技术发现鱼藤酮毒性作用造成的蛋白质组学改变。
     本研究成功地建立PD的细胞模型。0.5μM鱼藤酮处理PC12细胞72h,MTT检测细胞存活率下降到56.44±2.28%;Hoechst33342/PI双染细胞呈现亮蓝色的核边集、核固缩、核碎裂等凋亡特征,凋亡率达到了23.43±4.17%;HE染色细胞胞浆呈明显空泡变;蛋白质组学研究发现63个差异蛋白点,其中38个蛋白点显著上调、25个蛋白点显著下降。MALDI-TOF质谱鉴定出其中的17个差异蛋白点:明显上调的蛋白质包括vinculin、Grp75、TH、γ-enolase、SKRAP、ECH1、PAF-AH Ibα2、Hsp27、TPi1、GSTpi,明显下调的蛋白质包括ACO2、αtubulin-2、eIF4A1、MAP-EB1、SAE。这些变化的蛋白质可能参与鱼藤酮的毒性作用。
     本研究在鱼藤酮毒性处理的PD细胞模型中发现GSTpi、PAF-AH1bα2、Hsp27、vinculin、MAP-EB1、γ-enolase、Tpi1、ECH1、eIF4A、SAE及SKRAP蛋白表达的显著变化,这在以往的研究中尚未见报道,此外,还验证了ACO2在此模型中的变化。这些发现丰富了对这个领域的认识,为揭示PD等多因素参与的神经变性疾病病理过程的分子机制提供了大量新的线索。这些蛋白质有可能是PD病理机制中的关键分子和神经防护药物治疗的靶点。
Parkinson’s disease (PD) is also called“shaking palsy”. PD is one of the most common neurodegenerative diseases. It is characterized by selective and progressive degeneration of dopaminergic neurons and presence of Lewy bodies in the remaining neurons of the substantia nigra. To date, the cause and pathogenesis of PD remains unclarified, and the mechanisms underlying dopaminergic neuron degeneration are obscure. 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) can produce a syndrome which is clinically similar to PD. It has been hypothesized that environmental toxicants may contribute to the development of PD. Because MPTP has not been found in nature and only synthesized in limited quantities for the experimental research, it is unlikely that MPTP itself is the main cause of sporadic PD. Rotenone is a naturally occurring plant compound and a common insecticide used in vegetable gardens. It is also used to kill fish. Recently it has been demonstrated that rats administered subacute doses of rotenone develop biochemical, anatomical, and behavioral symptoms of PD. These results have renewed interest in the link between the pesticides exposure and the development of PD. Thus, rotenone treatment provides one of the best experimental models for PD research. The rotenone model may be an invaluable tool for investigating the environmental factors and the link between the cause and pathogenesis of PD. This model gives excellent experimental support to the environmental hypothesis of PD. Proteomics analysis of brain compartments may be useful to understand the complexity of PD, investigate disorders of the central nervous system and search for the corresponding early markers and screening the target of treatment in PD.
     In our experiment, the environmental toxin rotenone was used to create the cell model of PD. The PC12 cells are one of the best choices for the substitute of dopaminergic neuron. The aim of our studies was to further elucidate the toxic pathogenesis of rotenone by proteomics approach and provide a new insight for the cause and pathogeneis of PD.
     Part I The establishment of rotenone-treated PC12 cell model of Parkinson’s disease
     PC12 cells were treated with different concentrations of rotenone(0μM,0.1μM,0.5μM,1μM)for 24h, 48h and 72h, respectively. Cell morphological changes in PC12 cells were examined by phase–contrast microscopy. Cell viability was measured by MTT method. Cell apoptotic rate was assessed by Hoechst 33342/PI stains. The morphological results showed that the cell body became smaller and smoother, the processes of PC12 cells shortened or disappeared in a time- and concentration-dependent manner after being treated with rotenone for 24h, 48h and 72h, respectively. MTT showed cell viability decreased in 0.1μM rotenone-treated group afer 24h compared with control group ( p<0.05). The longer time past, the cell viability gradually descreased after rotenone treatment. Cell viability significantly decreased to 56.44±2.28% in 0.5μM rotenone-treated group at 72h. Hoechst33342/PI fluorescence staining showed that more apoptosis cells were found in rotenone-treated groups, the ratio of apoptosis increased significantly to 23.43±4.17% in 0.5μM rotenone-treated group at 72h. Cells shown condensed and fragmented nuclei demonstrated by fluorescent dyes, which are the characteristics of apoptosis. These results indicated that rotenone indeedly insulted PC12 cells. A widely accepted cell model of PD was established, which sets up a reliable basis for the study of rotenone mechanisms at proteomic level.
     Part II Proteomics study of toxic mechanism of rotenone
     Rotenone is a naturally occurring plant compound. As the special mitochondrial complex I inhibitor, rotenone can impair oxidation- phosphorylation of mitochondrial. The evidences from epidemiology and animal experiment indicate that rotenone is the candidate of cause factors for PD. However, since the rotenone model is a recently established model of PD, the molecular mechanisms underlying rotenone-induced insults of dopaminergic neurons are not well understood. Further exploring the toxic mechanism of rotenone at proteomic level may provide new clues to elucidate pathogenesis of PD.
     To explore the toxic mechanisms of rotenone, the goal of proteomics is to detect novel drug targets and diagnostic markers of PD.
     Proteins were extracted from control and 0.5uM rotenone-treated groups, respectively. The maps of proteins were set up by DIGE system. The altered protein spots were identified with MALDI-TOF MS and database searching.
     About 1740 protein spots were seen in 2D-DIGE images. The expressions of 63 proteins were significantly changed, of which 38 were up-regulated and 25 were down-regulated more than 20% in rotenone-treated PC12 cells compared with that in control. 17 protein spots were identified by mass spectrometry. The up-regulated proteins include vinculin, Grp75, TH,γ-enolase, SKRAP, ECH1, PAF-AH Ibα2, Hsp27, TPi1 and GSTpi. The down- regulated proteins include ACO2,αtubulin-2, eIF4A1, MAP-EB1 and SAE.
     The cytoskeleton protein,αtubulin-2, a member of the tubulin superfamily, is one of the components of microtubule and is required for the normal structure and normal fuction of microtubule. The microtubule associated protein EB1 (MAP-EB1)can regulate the assembly and functions of microtubules. Vinculin plays a pivotal role in cell adhesion, apoptosis and migration. Proteins with chaperone activity including Grp75 and Hsp27. Grp75 is a member of the heat shock/stress-response protein family. As a molecular chaperone, Hsp27 has neuroprotective effects via two actions: prevention of protein misfolding and acceleration of protein refolding and renaturation; inhibition of the apoptotic pathways within the cell. Hsp27 may be useful in the treatment of PD. ACO2 is a member of the Fe-S enzyme family. Its functions are involved TCA cycle and regulation of iron metabolism, which can contribute the pathogenesis of PD.γ-enolase and Tpi1 are both the key enzymes in glycolytic cycle. ECH1 is one of the key enzyme of the peroxisomalβ-oxidation cycle. The three enzyme (γ-enolase, Tpi1 and ECH1) regulate energy metabolism and may associate with the pathogenesis of PD. GSTpi has direct antioxidant activity and is involved in the metabolism of dopamine. GSTpi may be the key molecular of pathogenesis of PD. PAF-AH catalyzes PAF to inactivate it and has anti-inflammatory effects and anti-apoptotic actions. PAF-AH may affect the progress of neurodegeneration. eIF4A is the prototypic member of the DEAD box family of ATP-dependent RNA helicases and is involved in almost all aspects of RNA metabolism. eIF4A is capable of interacting directly with double-stranded RNA and contributes to unwind double-stranded RNA and RNA-protein complex. It is important in the initiating process of protein synthesis. SAE is a member of ubiqutin-like dynamic reversible key enzyme of posttranslational modification of proteins system. This system does not lead to degradate but instead to regulate protein-protein interactions, intracellular localization and protects some modifyed targets from ubiquitin-dependent degradation. SKRAP is associated with serine/threonine kinases receptor and regulates the cell differentiation, proliferation and apoptosis.
     In present study, the toxic mechanisms of rotenone were studied by DIGE and MS technology at proteomic level for the first time. It is the first time to have found the changed proteins of GSTpi, PAF-AH1bα2, hsp27, vinculin, MAP-EB1,γ-enolase, Tpi1, ECH1, eIF4A, SAE and SK-RAP in rotenone model of PD. The change of ACO2 is consistant with that from past. experiments in rotenone-treated cell model of PD. The alteration of Grp75 is different from that in other study in rotenone-treated cell model of PD. These changed protein may be invoved in the toxicity of rotenone and may be the candidate of diagnosis marker and the target of neuroprotecive treatment of PD.
引文
1. Dauer W, Przedborski S. Parkinson's disease: Mechanisms and models. Neuron, 2003; 39(6):889-909.
    2. 陈彪, 邹海强. 帕金森病分子生物学和临床研究进展. 中华神经杂志, 2002; 35(1):2-4.
    3. Goedert M. α-synuclein and neurodegenerative diseases. Nat Rev Neurosci, 2001; 2 (7):492-501.
    4. Gasser T. Genetics of Parkinson’s disease. J Neurol, 2001; 248(10):833-840.
    5. Langston JW, Ballard P, Tetrud JW, et al. Chronic Parkinsonism in humans due to a product of meperidine-analog synthesis. Science, 1983; 219(4587):979-980.
    6. Langston JW, Langston EB, Irwin I. MPTP-induced parkinsonism in human and non-human primates--clinical and experimental aspects. Acta Neurol Scand Suppl, 1984; 100:49-54.
    7. Przedborski S, Tieu K, Perier C, et al. MPTP as a mitochondrial neurotoxic model of Parkinson's disease. J Bioenerg Biomembr, 2004; 36(4): 375-379.
    8. Gorell JM, Johnson CC, Rybicki BA, et al. The risk of Parkinson's disease with exposure to pesticides, farming, well water, and rural living. Neurology, 1998; 50(5):1346-1350.
    9. Tuchen F. Jensen AA. Agricultural work and the risk of Parkinson’s disease in denmark, 1981-1993. Scand J Work Environ Health, 2000; 26(4):359-362.
    10. Ritz B, Yu F. Parkinson's disease mortality and pesticide exposure inCalifornia 1984-1994. Int J Epidemiol, 2000; 29(2): 323-329.
    11. Le Couteur DG, Muller M, Yang MC, et al. Age-environment and gene environment interactions in the pathogenesis of Parkinson’s disease. Rev Environ Health, 2002; 17(1):51-64.
    12. Helmuth L. Neuroscience.Pesticide causes Parkinson’s in rats. Science, 2000; 290(5494):l068.
    13. Priyadarshi A, Khuder SA, Schaub EA, et a1. Environmental risk factos and Parkinson’s disease: a meta analysis. Environ Res, 2001; 86(2):122-127.
    14. Ferrante RJ, Schulz JB, Kowall NW, et al. Systemic administration of rotenone produces selective damage in the striatum and globus pallidus, but not in the substantia nigra. Brain Res, 1997; 753(1):157-162.
    15. Betarbet R, Sherer TB, MacKenzie G, et al. Chronic systemic pesticide exposure reproduces features of Parkinson's disease. Nat Neurosci, 2000; 3 (12):1301-1306.
    16. Alam M, Schmidt WJ. Rotenone destroys dopaminergic neurons and induces parkinsonian symptoms in rats. Behav Brain Res, 2002; 136(1):317–324.
    17. Sherer TB, Kim JH, Betarbet R, et al. Subcutaneous rotenone exposure causes highly selective dopaminergic degeneration and alpha-synuclein aggregation. Exp Neurol, 2003; 179(1): 9-16.
    18. H(?)glinger GU, Féger J, Prigent A, et al. Chronic systemic complex I inhibition induces a hypokinetic multisystem degeneration in rats. J Neurochem, 2003; 84(3):491-502.
    19. Blackstock WP, Weir MP. Proteomics: quantitative and physical mapping of cellular proteins. Trends Biotechnol, 1999; 17(3): 121-127.
    20. Unlu M, Morgan ME, Minden JS. Difference gel electrophoresis: a single gel method for detecting changes in protein extracts. Electrophoresis, 1997; 18(11): 2071-2077.
    21. Das PC, McElroy WK, Cooper RL. Potential mechanisms responsible for chlorot-riazine induced alterations in catecholamines in pheochromocytoma (PC12) cells. Life Sci, 2003; 73(24):3123-3138.
    22. Isenberg JS, Klaunig JE. Role of the mitochondrial membrane permeability transition (MPT) in rotenone-induced apoptosis in liver cells. Toxicol Sci, 2000; 53(2):340–351.
    23. Beinert H, Kennedy MC. Aconitase, a two-faced protein: enzyme and iron regulatory factor. FASEB J, 1993; 7(15):1442–1449.
    24. Kim HY, LaVaute T, Iwai K, et al. Identification of a conserved and functional iron-responsive element in the 5`-untranslated region of mammalian mitochondrial aconitase. J Biol Chem, 1996; 271(39): 24226–24230.
    25. Schalinske KL, Chen OS, Eisenstein RS. Iron differentially stimulates translation of mitochondrial aconitase and ferritin mRNAs in mammalian cells. Implications for iron regulatory proteins as regulators of mitochondrial citrate utilization. J Biol Chem, 1998; 273(6):3740–3746.
    26. Liang LP, Ho YS, Patel M. Mitochondrial superoxide productionin kainate-induced hippocampal damage. Neuroscience, 2000; 101(3):563–570.
    27. Vivar JV, Kalyanaraman B, Kennedy MC. Mitochondrial aconitase is a source of hydroxyl radical. J Biol Chem, 2000; 275(19):14064–14069.
    28. Sofic E, Riederer P, Heinsen H, et al. Increased iron (III) and total ironcontent in post mortem substantia nigra of parkinsonian brain. J Neural Transm, 1988; 74(3):199–205.
    29. Kaur D, Yantiri F, Rajagopalan S, et al. Genetic or pharmacological iron chelation prevents MPTP-induced neurotoxicity in vivo: A novel therapy for Parkinson’s disease. Neuron, 2003; 37(6):899–909.
    30. Liang L, Patel M. Iron-sulfur enzyme mediated mitochondrial superoxide toxicity in experiential Parkinson’s disease. J Neurochem, 2004; 90(5):1076–1084.
    31. Rouault TA, Klausner RD. Iron-sulfur clusters as biosensors of oxidants and iron. Trends Biochem Sci, 1996; 21(5):174–177.
    32. Tabrizi SJ, Cleeter MW, Xuereb J, et al. Biochemical abnormalities and excitotoxicity in Huntington’s disease brain. Ann Neurol, 1999; 45(1):25–32.
    33. Albers DS, Swerdlow RH, Manfredi G, et al. Further evidence for mitochondrial dysfunction in progressive supranuclear palsy. Exp Neurol, 2001; 168(1):196–198.
    34. Patel M. Mitochondrial dysfunction and oxidative stress: Cause and consequence of epileptic seizures. Free Radic Biol Med, 2004; 37(12): 1951–1962.
    35. Cho SG, Lee YH, Park HS, et al. Glutathione S-transferase mu modulates the stress-activated signals by suppressing apoptosis signal-regulating kinase 1. J Biol Chem, 2001; 276(16):12749-12755.
    36. Beiswanger CM, Diegmann MH, Novak RF, et al. Developmental changes in the cellular distribution of glutathione and glutathione S-transferases in the murine nervous system. Neurotoxicology, 1995; 16(3):425–440.
    37. Awasthi YC, Sharma R, Singhal SS. Human glutathione S-transferases. Int J Biochem, 1994; 26(3):295–308.
    38. Oakley AJ, Lo Bello M, Nuccetelli M, et al. The ligandin (non-substrate) binding site of human pi class glutathione transferase is located in the electrophile binding site (H-site). J Mol Biol, 1999; 291(4):913 - 926.
    39. Olanow CW, Tatton WG. Etiology and pathogenesis of Parkinson’s disease. Annu Rev Neurosci, 1999; 22:123–144.
    40. Beal MF. Mitochondria, oxidative damage, and inflammation in Parkinson's disease. Ann NY Acad Sci, 2003; 991:120–131.
    41. Peng J, Andersen JK. The role of c-Jun N-terminal kinase (JNK) in Parkinson's disease. IUBMB Life, 2003; 55(4-5):267–271.
    42. Kaur D, Andersen J. Does cellular iron dysregulation play a causative role in Parkinson's disease? Ageing Res Rev, 2004; 3(3):327–343.
    43. Di Ilio C, Sacchetta P, Iannarelli V, et al. Binding of pesticides to alpha, mu and pi class glutathione transferase. Toxicol Lett, 1995; 76(2):173–177.
    44. Baez S, Segura-Aguilar J, Widersten M, et al. Glutathione transferases catalyse the detoxication of oxidized metabolites (o-quinones) of catecholamines and may serve as an antioxidant system preventing degenerative cellular processes. Biochem J, 1997; 324(Pt 1): 25–28.
    45. Wang T, Arifoglu P, Ronai Z, et al. Glutathione S-transferase P1-1 (GSTP1-1) inhibits c-Jun N-terminal kinase (JNK1) signaling through interaction with the C terminus. J Biol Chem, 2001; 276(24):20999–21003.
    46. Zimniak P, Nanduri B, Pikula S, et al. Naturally occurring human glutathione S-transferase GSTP1-1 isoforms with isoleucine and valine in position 104 differ in enzymatic properties. Eur J Biochem, 1994; 224(3):893–899.
    47. Menegon A, Board PG, Blackburn AC, et al. Parkinson’s disease, pesticides, and glutathione transferase polymorphisms. Lancet, 1998; 352(9137):1344-1346.
    48. Ha X, Xia H, Srivastava SK, et al. Activity of four allelic forms of glutathione S-transferase hGSTP1-1 for diol epoxides of polycyclic aromatic hydrocarbons. Biochem Biophys Res Commun, 1997; 238(2): 397–402.
    49. Schaloske RH, Dennis EA. The phospholipase A2 superfamily and its group numbering system. Biochim Biophys Acta, 2006; 1761(11):1246-1259.
    50. Hattori M, Arai H, Inoue K. Purification and characterization of bovine brain platelet-activating factor acetylhydrolase. J Biol Chem, 1993; 268(25):18748–18753.
    51. Ho YS, Swenson L, Derewenda U, et al. Brain acetylhydrolase that inactivates platelet-activating factor is a G-protein-like trimer. Nature, 1997; 385(6611):89–93.
    52. Hattori M, Adachi H, Tsujimoto M, et al. The catalytic subunit of bovine brain platelet-activating factor acetylhydrolase is a novel type of serine esterase. J Biol Chem, 1994; 269(37):23150–23155.
    53. Adachi H, Tsujimoto M, Hattori M, et al. cDNA cloning of human cytosolic platelet-activating factor acetylhydrolase gamma-subunit and its mRNA expression in human tissues. Biochem Biophys Res Commun, 1995; 214(1):180–187.
    54. Albrecht U, Abu-Issa R, Ratz B, et al. Platelet-activating factor acetylhydrolase expression and activity suggest a link between neuronalmigration and platelet-activating factor. Dev Biol, 1996; 180(2):579–593.
    55. Watanabe M, Aoki J, Manya H, et al. Molecular cloning of cDNAs encoding alpha1, alpha2, and beta subunits of rat brain platelet-activating factor acetylhydrolase. Biochim Biophys Acta, 1998; 1401(1):73–79.
    56. Manya H, Aoki J, Kato H, et al. Biochemical characterization of various catalytic complexes of the brain platelet-activating factor acetylhydrolase. J Biol Chem, 1999; 274(45):31827-31832.
    57. Tjoelker LW, Stafforini DM. Platelet-activating factor acetylhydrolases in health and disease. Biochim Biophys Acta, 2000; 1488(1-2):102-123.
    58. Bazan NG. The neuromessenger platelet-activating factor in plasticity and neurodegeneration. Prog Brain Res, 1998; 118:281-291.
    59. Bennett SA, Chen J, Pappas BA, et al. Platelet activating factor receptor expression is associated with neuronal apoptosis in an in vivo model of excitotoxicity. Cell Death Differ, 1998; 5(10):867-875.
    60. Bonin F, Ryan SD, Migahed L, et al. Anti-apoptotic actions of the platelet-activating factor acetylhydrolase I alpha2 catalytic subunit. J Biol Chem, 2004; 279(50):52425-52436.
    61. Kappe′ G, Franck E, Verschuure P, et al. The human genome encodes 10 α-crystallin-related heat shock proteins: HspB1-10. Cell Stress Chaperones, 2003; 8(1):53–61.
    62. Plumier JC, Hopkins DA, Robertson HA, et al. Constitutive expression of the 27-kDa heat shock protein (Hsp27) in sensory and motor neurons of the rat nervous system. J Comp Neurol, 1997; 384(3):409–428.
    63. Plumier JC, Armstrong JN, Landry J, et al. Expression of the 27,000 mol. wt heat shock protein following kainic acid-induced status epilepticus in therat. Neuroscience, 1996; 75(3):849–856.
    64. Currie RW, Ellison JA, White RF, et al. Benign focal ischemic preconditioning induces neuronal Hsp70 and prolonged astrogliosis with expression of Hsp27. Brain Res, 2000; 863(1-2):169–181.
    65. Krueger-Naug AM, Emsley JG, Myers TL, et al. Injury to retinal ganglion cells induces expression of the small heat shock protein hsp27 in the rat visual system. Neuroscience, 2002; 110(4):653–665.
    66. Calabrese V, Scapagnini G, Ravagna A, et al. Increased expression of heat shock proteins in rat brain during aging: relationship with mitochondrial function and glutathione redox state. Mech Ageing Deve, 2004; 125(4):325–335.
    67. Pringle AK, Thomas SJ, Signorelli F, et al. Ischaemic pre-conditioning in organotypic hippocampal slice cultures is inversely correlated to the induction of the 72 kDa heat shock protein (HSP72). Brain Res, 2000; 867(1-2):62–69.
    68. Akbar MT, Lundber AMC, Vidyadaran S, et al. The neuroprotective effects of heat shock protein 27 overexpression in transgenic animals against kainate-induced seizures and hippocampal cell death. J Biol Chem, 2003; 278(22):19956–19965.
    69. Kwong JM, Lam TT, Caprioli J. Hyperthermic pre-conditioning protects retinal neurons from N-methyl-Daspartate (NMDA)-induced apoptosis in rat. Brain Res, 2003; 970(1-2):119–130.
    70. Samali A, Cotter TG. Heat shock proteins increases resistance to apoptosis. Exp Cell Res, 1996; 223(1):163–170.
    71. Mehlen P, Preville X, Chareyron P, et al. Constitutive expression of humanhsp27, Drosophila hsp27, or human alpha B-crystallin confers resistance to TNF- and oxidative stress-induced cytotoxicity in stably transfected murine L929 fibroblasts. J Immun, 1995; 154(1):363–374.
    72. Bukau B, Weissman J, Horwich A. Molecular chaperones and protein quality control. Cell, 2006; 125 (3): 443–451.
    73. Gorman AM, Szegezdi E, Quigney DJ, et al. Hsp27 inhibits 6-hydroxydopamine-induced cytochrome c release and apoptosis in PC12 cells. Biochem Biophys Res Commun, 2005; 327(3):801-810.
    74. Benn SC, Perrelet D, Kato AC, et al. Hsp27 Upregulation and phosphorylation is required for injured sensory and motor neuron survival. Neuron, 2002; 36(1):45–56.
    75. Kalwy S, Akbar MT, Coffin S, et al. Heat shock protein 27 delivered via a herpes simplex virus vector can protect neurones of the hippocampus against kainic acid-induced cell loss. Mol Brain Res, 2003; 111(1-2):91–103.
    76. Zourlidou A, Payne Smith M, Latchman DS. Hsp27 but not Hsp70 has a potent protective effect against α-synuclein-induced cell death in mammalian neuronal cells. J Neurochem, 2004; 88(6):1439–1448.
    77. Wyttenbach A, Sauvageot O, Carmichael J, et al. Heat shock protein 27 prevents cellular polyglutamine toxicity and suppresses the increase of reactive oxygen species caused by huntingtin. Hum Mol Genet, 2002; 11(9):1137–1151.
    78. Massa SM, Longo FM, Zuo J, et al. Cloning of rat grp75, an hsp70 family member, and its expression in normal and ischemic brain. J Neurosci Res, 1995; 40(6): 807-819.
    79. Ran Q, Wadhwa R, Kawai R, et al. Extramitochondrial localization of mortalin/mthsp70/PBP74/GRP75. Biochem Biophys Res Commun, 2000; 275(1): 174–179.
    80. Ryan MT, Naylor DJ, Hoj PB, et al. The role of molecular chaperones in mitochondrial protein import and folding. Int Rev Cytol, 1997; 174:127-193.
    81. Savel'ev AS, Novikova LA, Kovaleva IE, et al. ATP-dependent proteolysis in mitochondria. m-AAA protease and PIM1 protease exert overlapping substrate specificities and cooperate with the mtHsp70 system. J Biol Chem, 1998; 273(32):20596-20602.
    82. Jin J, Hulette C, Wang Y, et al. Proteomic identification of a stress protein, mortalin/mthsp70/GRP75: relevance to Parkinson disease. Mol Cell Proteomics, 2006; 5(7):1193-1204.
    83. Janssen ME, Kim E, Liu H, et al. Three-dimensional structure of vinculin bound to actin filaments. Mol Cell, 2006; 21(2):271-281.
    84. Rodriguez-Fernandez JL, Geiger B, Salomon D, et al. Suppression of tumorigenicity in transformed cells after transfection with vinculin Cdna. J Cell Biol, 1992; 119(2):427-438.
    85. Subauste MC, Pertz O, Adamson ED, et al. Vinculin modulation of paxillin-FAK interactions regulates ERK to control survival and motility. J Cell Biol, 2004; 165(3):371-381.
    86. Mailliot C, Podevin-Dimster V, Rosenthal RE, et al. Rapid tau protein dephosphorylation and differential rephosphorylation during cardiac arrest-induced cerebral ischemia and reperfusion. J Cereb Blood Flow Metab, 2000; 20(3):543-549.
    87. Olanow CW, Perl DP, DeMartino GN, et al. Lewy-body formation is anaggresome-related process: a hypothesis. Lancet Neurol, 2004; 3(8):496-503.
    88. Alim MA, Hossain MS, Arima K. Tubulin seeds alpha-synuclein fibril formation. J Biol Chem, 2002; 277(3):2112-2117.
    89. Lee HJ, Khoshaghideh F, Lee S, et al. Impairment of microtubule-dependent trafficking by overexpression of alpha-synuclein. Eur J Neurosci, 2006; 24(11):3153-3162.
    90. Crosby AH. Disruption of cellular transport: a common cause of neurodegeneration? Lancet Neurol, 2003; 2(5):311-316.
    91. Diaz-Corrales FJ, Asanuma M, Miyazaki I, et al. Rotenone induces aggregation of gamma-tubulin protein and subsequent disorganization of the centrosome: relevance to formation of inclusion bodies and neurodegeneration. Neuroscience, 2005; 133(1):117-135.
    92. Feng J. Microtubule: a common target for parkin and Parkinson's disease toxins. Neuroscientist, 2006; 12(6):469-476.
    93. Su LK, Burrell M, Hill DE, et al. APC binds to the novel protein EB1. Cancer Res, 1995; 55(14):2972–2977.
    94. Berrueta L, Tirnauer JS, Schuyler SC, et al. The APC-associated protein EB1 associates with components of the dynactin complex and cytoplasmic dynein intermediate chain. Curr Biol, 1999; 9(8):425-428.
    95. Askham JM, Vaughan KT, Goodson HV, et al. Evidence that an interaction between EB1 and p150 (Glued) is required for the formation and maintenance of a radial microtubule array anchored at the centrosome. Mol Biol Cell, 2002; 13(10):3627-3245.
    96. Hayashi I, Ikura M. Crystal structure of the amino-terminal microtubule-binding domain of end-binding protein 1 (EB1). J Biol Chem, 2003; 278(38):36430–36434.
    97. Slep KC, Rogers SL, Elliott SL, et al. Structural determinants for EB1-mediated recruitment of APC and spectraplakins to the microtubule plus end. J Cell Biol, 2005; 168(4):587–598.
    98. Keller A, Berod A, Dussaillant M, et al. Coexpression of alpha and gamma enolase genes in neurons of adult rat brain. J Neurosci Res, 1994; 38(5):493–504.
    99. Schonberger SJ, Edgar PF, Kydd R, et al. Proteomic analysis of the brain in Alzheimer's disease: molecular phenotype of a complex disease process. Proteomics, 2001; 1(12):1519-1528.
    100.杨国锋, 王鲁宁, 何思志等. Pick 病的蛋白质组研究. 中华神经科杂志, 2006; 39(2):96-100.
    101.Castegna A, Thongboonkerd V, Klein JB, et al. Proteomic identification of nitrated proteins in Alzheimer's disease brain. J Neurochem, 2003; 85(6):1394-1401.
    102.Gautam A,Hung WL. Enoyl-coA hydratase: reaction, mechanism, and inhibition. Bioorg Med Chem, 2003; 11(1):9-20.
    103.Sato S, Nomura CT, Abe H, et al. Poly[(R)-3-hydroxybutyrate] formation in Escherichia coli from glucose through an enoyl-CoA hydratase-mediated pathway. J Biosci Bioengi, 2007; 103(1):38-44.
    104.Yang GF, Wang LN, Ji JG, et al. A preliminary proteomic analysis of tauopathies. Zhonghua Nei Ke Za Zhi, 2005; 44(5):374-377.
    105.Linder P, Lasko PF, Ashburner M, et al. Birth of the D-E-A-D box. Nature,1989; 337(6203):121–122.
    106.de la Cruz J, Kressler D, Linder P. Unwinding RNA in Saccharomyces cerevisiae: DEAD-box proteins and related families. Trends Biochem Sci, 1999; 24(5):192–198.
    107.Luking A, Stahl U, Schmidt U. The protein family of RNA helicases. Crit Rev Biochem Mol Biol, 1998; 33(4):259–296.
    108.Song P, Zhang JN, Hu JR, et al. cDNA Cloning expression and ATPase assay of a putative eIF-4A of plasmodium cynomolgi. Chinese J Biochem Mol Biol, 2004; 20 (3):294–304.
    109.Yeh ET, Gong L, Kamitani T. Ubiquitin-like proteins: new wines in new bottles. Gene, 2000; 248(1-2):1–14.
    110.Boggio R, Chiocca S. Gam1 and the SUMO pathway. Cell Cycle, 2005; 4(4):533-535.
    111.Hochstrasser M. Evolution and function of ubiquitin-like protein-conjugation systems. Nat Cell Biol, 2000; 2(8):E153–E157.
    112.Desterro JM, Rodriguez MS, Kemp GD, et al. Identification of the enzyme required for activation of the small ubiquitin-like protein SUMO-1. J Biol Chem, 1999; 274(15): 10618–10624.
    113.Wilson VG, Rangasamy D. Intracellular targeting of proteins by sumoylation. Exp Cell Res, 2001; 271(1):57–65.
    114.Cross TG, Scheel-Tollner D, Henriquez NV, et al. Serine/threonine protein kinases and apoptosis. Exp Cell Res, 2000; 256(1):34 – 41.
    1. Dauer W, Przedborski S. Parkinson's disease: mechanisms and models. Neuron, 2003; 39(6):889-909.
    2. Gasser T. Genetics of Parkinson's disease. J Neurol, 2001; 248(10):833-840.
    3. Langston JW, Ballard P, Tetrud JW, et al. Chronic Parkinsonism inhumans due to a product of meperidine-analog synthesis. Science, 1983; 219(4587):979-980.
    4. Schober A. Classic toxin-induced animal models of Parkinson’s disease: 6-OHDA and MPTP. Cell Tissue Res, 2004; 318(1):215-224.
    5. Gorell JM, Johnson CC, Rybicki BA, et al. The risk of Parkinson's disease with exposure to pesticides, farming, well water, and rural living. Neurology, 1998; 50(5):1346-1350.
    6. Priyadarshi A, Khuder SA, Schaub EA, et al. A meta-analysis of Parkinson's disease and exposure to pesticides. Neurotoxicology, 2000; 21 (4):435-440.
    7. Ritz B, Yu F. Parkinson's disease mortality and pesticide exposure in California 1984-1994. Int J Epidemiol, 2000; 29 (2): 323-329.
    8. 杨仕平, 黄继光, 徐汉虹等. 鱼藤酮的晶体结构及生物活性. 上海师范大学学报, 2003; 32(3):47-51.
    9. Talpade DJ, Greene JG, Higgins DSJ, et al. In vivo labeling of mitochondrial complex I (NADH: ubiquinone oxidoreductase) in rat brain using [3H] dihydrorotenone. J Neurochem, 2000; 75(6):2611–2621.
    10. Schuler F, Casida JE. Functional coupling of PSST and ND1 subunits in NADH: ubiquinone oxidoreductase established by photoaffinity labeling. Biochim Biophys Acta, 2001; 1506(1):79–87.
    11. Stern ME, Dulaney SB, Gruber L, et al. The epidemiology of Parkinson's disease. Acase–control study of young-onset and old-onset patients. Arch Neurol, 1991; 48(9):903–907.
    12. Semchuk KM, Love EJ. Effects of agricultural work and other proxy-derived case-control data on Parkinson's disease risk estimates. AmJ Epidemiol, 1995; 141(8):747-754.
    13. 菲琳. 帕金森病可能与杀虫剂有关. 国外医学情报, 2002; 07.
    14. Betarbet R, Sherer TB, MacKenzie G, et al. Chronic systemic pesticide exposure reproduces features of Parkinson's disease. Nat Neurosci, 2000; 3 (12): 1301- 1306.
    15. Alam, M. Schmidt, WJ. Rotenone destroys dopaminergic neurons and induces parkinsonian symptoms in rats. Behav Brain Res, 2002; 136(1):317-318.
    16. Sherer TB, Kim JH, Betarbet R, et al. Subcutaneous rotenone exposure causes highly selective dopaminergic degeneration and alpha-synuclein aggregation. Exp Neurol, 2003; 179(1):9-16.
    17. H?glinger GU, Féger J, Prigent A, et al. Chronic systemic complex I inhibition induces a hypokinetic multisystem degeneration in rats. J Neurochem, 2003; 84(3):491-502.
    18. 冯媛, 梁直厚, 王涛等. 鱼藤酮帕金森病大鼠模型建立的研究. Stroke Nerv Dis, 2004; 11(6):374-377.
    19. 赵喜林,顾振纶,秦正红. 长期低剂量皮下注射鱼藤酮制作大鼠帕金森病模型. 中国药理学通报, 2005; 21(10):1274-1277.
    20. Hatefi Y. The mitochondrial electron transport and oxidative phosphorylation system. Annu Rev Biochem, 1985; 54: 1015-1069.
    21. Schapira AH, Cooper JM, Dexter D, et al. Mitochondrial complex I deficiency in Parkinson's disease. J Neurochem, 1990; 54 (3): 823-827.
    22. Dawson TM, Dawson VL. Molecular Pathways of Neurodegeneration in Parkinson'sDisease. Science, 2003; 302(5646):819-822.
    23. van der Walt JM, Nicodemus KK, Martin ER, et al. MitochondrialPolymorphisms Significantly Reduce the Risk of Parkinson Disease. Am J Hum Gene, 2003; 72(4): 804-811.
    24. Seaton TA, Cooper JM, Schapira AHV. Free radical scavengers protect dopaminergic cell lines from apoptosisinduced by complex I inhibitors. Brain Res, 1997; 777(1-2):110–118.
    25. Sherer TB, Betarbet R, Testa CM, et al. Mechanism of toxicity in rotenone models of Parkinson's disease. J Neurosci, 2003b; 23(34):10756-10764.
    26. Bonsi P, Calabresi P, De Persis C, et al. Early ionic and membrane potential changes caused by the pesticide rotenone in striatal cholinergic interneurons. Exp Neurol, 2004; 185(1):169-181.
    27. Good PF, Hsu A, Werner P, et al. Protein nitration in Parkinson's disease. J Neuropathol Exp Neurol, 1998; 57(4):338-342.
    28. Sipos I, Tretter L, Adam-ViziV. Quantitative relationship between inhibition of respiratory complexes and formation of reactive oxygen species in isolated nerve terminals. J Neurochem, 2003; 84 (1):112-118.
    29. Bashkatova V, Alam M, VaninA, et al. Chronic administration of rotenone increases levels of nitric oxide and lipid peroxidation products in rat brain. Exp Neurol, 2004; 186(2):235-241.
    30. Ueda S, Masutani H, Nakamura H, et al. Redox control of cell death. Antioxid Redox Signal, 2002; 4(3):405–414.
    31. Anglade P, Vyas S, Javoy-Agid F, et al. Apoptosis and autophagy in nigral neurons of patients with Parkinson's disease. Histol Histopathol, 1997; 12 (1): 25-31.
    32. Tompkins MM, Basgall EJ, Zamrini E, et al. Apoptotic-like changes in Lewy-body-associated disorders and normal aging in substantia nigralneurons. Am J Pathol, 1997; 150(1):119-131.
    33. Tatton WG, Chalmers-Redman R, Brown D, et al. Apoptosis in Parkinson's disease: signals for neuronal degradation. Ann Neurol, 2003; 53 (Suppl 3): S61-70.
    34. Isenberg JS, Klaunig JE. Role of the mitochondrial membrane permeability transition (MPT) in rotenone-induced apoptosis in liver cells. Toxicol Sci, 2000; 53(2):340–351.
    35. Molina-Jiménez MF, Sánchez-Reus MI, Andres D, et al. Neuroprotective effect of fraxetin and myricetin against rotenone-induced apoptosis in neuroblastoma cells. Brain Res, 2004; 1009(1-2): 9-16.
    36. Seaton TA, Cooper JM, Schapira AH. Cyclosporin inhibition of apoptosis induced by mitochondrial complex I toxins. Brain Res, 1998; 809(1):12-17.
    37. Newhouse K, Hsuan SL, Chang SH, et al. Rotenone-induced apoptosis is mediated by p38 and JNK MAP kinases in human dopaminergic SH-SY5Y cells. Toxicol Sci, 2004; 79(1):137-146.
    38. King TD, Bijur GN, Jope RS. Caspase-3 activation induced by inhibition of mitochondrial complex I is facilitated by glycogen synthase kinase-3beta and attenuated by lithium. Brain Res, 2001; 919(1):106-114.
    39. Ryu EJ, Harding HP, Angelastro JM, et al. Endoplasmic reticulum stress and the unfolded protein response in cellular models of Parkinson's disease. J Neurosci, 2002; 22 (24):10690-10698.
    40. Ahmadi FA, Linseman DA, Grammatopoulos TN, et al. The pesticide rotenone induces caspase-3-mediated apoptosis in ventral mesencephalic dopaminergic neurons. J Neurochem, 2003; 87(4):914- 921.
    41. Stokes AH, Hastings TG, Vrana KE. Cytotoxic and genotoxic potential of dopamine. J Neurosci Res, 1999; 55(6):659-665.
    42. Burke WJ, Li SW, Chung HD, et al. Neurotoxicity of MAO metablites of catecholamine neurotransmitters: role in neurodegenerative diseases. Neurotoxicology, 2004; 25(1-2):101-115.
    43. Ito Y, Fujita M, Shimada S, et al. Comparison between the decrease of dopamine transporter and that of L-DOPA uptake for detection of ealy to aduanced stage of parkinson disease in animal models. Synapse, 1999; 31(3):178-185.
    44. Sakka N, Sawada H, Izumi Y, et al. Dopamine is involved in selectivity of dopaminergic neuronal death by rotenone. Neuroreport, 2003; 14(18): 2425-2428.
    45. Thiffault C, Langston JW, Di Monte DA. Increased striatal dopamine turnover following acute administration of rotenone to mice. Brain Res, 2000, 885(2):283-288.
    46. Burke WJ, Li SW, Williams EA, et al. 3,4-Dihydroxyphenylacetaldehyde is the toxic dopamine metabolite in vivo: implications for Parkinson’s disease pathogenesis. Brain Res, 2003; 989(2): 205-213.
    47. McGeer PL, McGeer EG. Glial cell reactions in neurodegenerative diseases: pathophysiology and therapeutic interventions. Alzheimer Dis Assoc Disord, 1998; 12(Suppl 2): S1-6.
    48. Beal MF. Mitochondria, oxidative damage, and inflammation in Parkinson's disease. Ann N Y Acad Sci, 2003; 991: 120-131.
    49. Sherer TB, Betarbet R, Kim JH, et al. Selective microglial activation in the rat rotenone model of Parkinson's disease. Neurosci Lett, 2003; 341(2):87-90.
    50. Gao HM, Hong JS, Zhang W, et al. Distinct role for microglia in rotenone-induced degeneration of dopaminergic neurons. J Neurosci, 2002; 22 (3):782-790.
    51. Gao HM, Liu B, Hong JS. Critical role for microglial NADPH oxidase in rotenone-induced degeneration of dopaminergic neurons. J Neurosci, 2003; 23(15):6181-6187.
    52. Gao HM, Hong JS, Zhang W, et al. Synergistic dopaminergic neurotoxicity of the pesticide rotenone and inflammogen lipopolysaccharide: relevance to the etiology of Parkinson’s disease. J Neurosci, 2003; 23(4):1228-1236.
    53. Marey-Semper I, Gelman M, Levi-Strauss M. A selective toxicity toward cultured mesencephalic dopaminergic neurons is induced by the synergistic effects of energetic metabolism impairment and NMDA receptor activation. J Neurosci, 1995; 15 (9): 5912 - 5918.
    54. Shamoto-Nagai M, Maruyama W, Kato Y. An inhibitor of mitochondrial complex I, rotenone, inactivates proteasome by oxidative modification and induces aggregation of oxidized proteins in SH-SY5Y cells. J Neurosci Res, 2003; 74 (4):589-597.
    55. Uversky VN, Li J, Fink AL. Pesticides directly accelerate the rate of alpha- synuclin fibril formation: a possible factor in Parkinson’s disease. FEBS Lett, 2001; 500(3):105-108.
    1. Elkon H, Melamed E, Offen D. 6-Hydroxydopamine increases ubiquitin-conjugate and protein degradation : implications for the pathogenesis of Parkinson’s disease. Cell Mol Neurobiol, 2001; 21(6):771-781
    2. Jeon BS, Jackson-Lewis, Burke RE. 6-Hydroxydopamine lesions of the rat substantia nigra: Time course and morphology of cell death. Neurodegeneraton, 1995; 4(2):131-137
    3. Praedborski S, Levivier M, Jiang H, et al. Dose-dependent lesions of the dopaminergic nigrostriatal pathway induced by intrastriatal injection of 6-hydroxydopamine. Neuroscience, 1995; 67(3):631-647.
    4. Bjorklund LM, Sanchez-Pernaute R, Chung S, et al. Embryonic stem cells develop into functional dopaminergic neurons after transplantation in a Parkinson rat model. Proc Natl Acad Sci USA, 2002; 99(4):2344-2349.
    5. Fabre E, Monserrat J, Herrero A, et al. Effect of MPTP on brain mitochondrial H2O2 and ATP production and on dopamine and DOPAC inthe striatum. J Physiol Biochem, 1999; 55(4):325-331.
    6. Viswanath V, Wu Y, Boonplueang R, et al. Caspase-9 activation results in downstream caspase-8 activation and bid cleavage in 1-methyl-4phenyl-1,2,3,6-tetrahydropyridine-induced Parkinson’s disease. J Neurosci, 2001; 21(24):9519-9528.
    7. Vila M, Jackson-Lewis V, Vukosavic S, et al. Bax ablation prevents dopaminergic neurodegeneration in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson’s disease. Proc Natl Acad Sci USA, 2001; 98(5):2837-2842.
    8. Kopin IJ, Markey SP. MPTP toxicity: implications for research in Parkinson’s disease. Annu Rew Neurosci, 1988; 11:81-96.
    9. Kitamura Y, Taniguchi T, et al. Neuroprotective mechanisms of antiparkinsonian dopamin D2-receptor subfamily agonists. Neurochem Res, 2003; 28(7):1035-1040.
    10. McCormack AL, Thiruchelvam M, Manning-Bog AB, et al. Environmental risk factors and Parkinson’s disease: selective degeneration of nigral dopaminergic neurons caused by the herbicide paraquat. Neurobiol Dis, 2002; 10(2):119-127.
    11. Betarbet R, Sherer TB, MacKenzie, et al. Chronic systemic pesticide exposure reproduces features of Parkinson’s disease. Nat Neurosci, 2000; 3(12):1301-1306.
    12. Kevin SP, McNaught, Daniel P, et al. Systemic exposure to proteasome inhibitors cause a progressive model of Parkinson’s disease. Ann Neurol, 2004; 56(1): 149-162.
    13. Giasson BI, Duda JE, Quinn, SM. Neuronal alpha-synuclinopathy withsevere movement disorder in mice expressing A53T human aspha-synuclein. Neuron, 2002; 34(4):521-533.
    14. Feany MB, Bender WW. A Drosophila model of Parkinson’s disease. Nature, 2000; 404(6776):394-398.
    15. Kirik D, Rosenblad C, Burger C, et al. Parkinson-like neurodegeneration induced by targetd overexpression of alpha-synuclein in the nigrostriatal system. J Neurosci, 2002; 22(7):2780-2791.
    16. Pendleton RG, Parvez F, Sayed M, et al. Effects of pharmacological agents upon a transgenic model of Parkinson’s disease in Drosophila melanogaster. J Pharmacol Exp Ther, 2002; 300(1):91-96.
    17. Hsu LJ, Sagara Y, Arroyo A, et al. alpha-synuclein promotes mitochondrial deficit and oxidative stress. Am J Pathol, 2000; 157(2):401–410.
    18. Lotharius J, Brundin P. Impaired dopamine storage resulting from alpha-synuclein mutations may contribute to the pathogenesis of Parkinson’s disease. Hum Mol Genet, 2002; 11(20):2395–2407.
    19. Sawada H, Shimohama S, Kawamura T, et al. Methylphenylpyridiumion (MPP+) enhances glutamate-induced cytotoxicity against dopaminergic neurons in cultured rat mesencephalon. J Neurosci Res, 1996; 43(1):55-62.
    20. Ferrante RJ, Hantraye PB, Brouillet E, et al. Increased nitrotyrosine immunoreactvity in substantia nigra neurons in MPTP treated baboons is blocked by inhibition of neuronal nitric oxide synthase. Brain Res, 1999; 823(1-2):177-182.
    21. Sawada H, Ibi M, Kihara T, et al. Estradiol protects dopaminergic neurons in a MPP+ Parkinson’s disease model. Neuropharmacology, 2002; 42 (8):1056-1064.

© 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