PrP 106-126对分化PC12细胞的毒性作用研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
  • 英文题名:Cytotoxic Effects of Differentiated PC12 Cell Infected by Prion Protein 106-126 Peptide
  • 作者:张东威
  • 论文级别:博士
  • 学科专业名称:神经病学
  • 学位年度:2005
  • 导师:赵节绪
  • 学科代码:100204
  • 学位授予单位:吉林大学
  • 论文提交日期:2005-04-01
  • 答辩委员会主席:黄佰渠
摘要
本研究通过建立朊蛋白106-126 肽段(PrP106-126)感染分化PC12细胞模型,利用流式细胞术、激光共聚焦显微镜技术、免疫组化以及蛋白质印记等技术从形态和功能两个方面对PrP106-126在分化PC12细胞上的毒性作用、具体的作用机制以及该方法能否作为朊蛋白病的发病模型进行了研究,观察了从早期出现的氧化应激到最终细胞凋亡的过程。朊蛋白病是一类由具有传染性的朊蛋白(Prion protein,PrP)为病原体所致的散发的人畜共患的中枢神经系统变性疾病,是一组既有传染性又有遗传性的变性疾病。朊蛋白疾病的核心是朊蛋白的构型由正常的PrPC向致病的PrPSC的转化,其共同的病理学特征是脑的海绵状变性。但是在发病机制上,PrPSC在脑内沉积如何引起继发的病理变化,目前尚不清楚,这也是朊蛋白病一直缺乏针对性治疗的原因之一。鉴于PrP106-126 具有许多PrPSC的生物学特性,通过建立PrP106-126 感染分化PC12 细胞模型,本研究发现,PrP106-126 对分化PC12 细胞具有确切的毒性作用,细胞感染PrP106-126 后较早出现氧化应激现象,并且氧化应激持续存在,随后出现细胞内钙离子浓度增加、线粒体膜电位的下降、Ca~(2+)ATP 酶活性降低,细胞能量代谢受到严重干扰,最终导致细胞凋亡,Bcl-2/Bax 系统稳态的变化和Caspase-3 的活化参与了诱导凋亡的过程。本研究认为PrP106-126 感染分化PC12 细胞是研究朊蛋白病的高效模型,PrP106-126 的毒性作用是通过持续的氧化应激、干扰细胞的能量代谢、钙超载、最终诱导宿主细胞的凋亡来实现的,并且氧化应激处于始动与核心的地位,提示从抗氧化损伤角度着手筛选药物可能是在朊蛋白病治疗上的一种值得尝试的方法。
It is believed that prion diseases are infectious, inherited, sporadic spongiform degeneration encephalopathies caused by prion protein conformational change. These disorders include scrapie of sheep and goats, bovine spongiform encephalopathy (BSE) of cattle, and Creutzfeldt-Jakob disease (CJD), Gerstmann-Straussler-Scheinker disease (GSS) and fatal familial insomnia (FFI) of humans. CJD is a rare disease that occurs in three forms: sporadic, genetic, and iatrogenic. More recently, a new variant of CJD (nvCJD) has been reported. All forms of transmissible spongiform encephalopathyes (TSEs) are characterized by spongiform degeneration of the brain, reactive gliosis, and neuronal loss. They are associated with the accumulation of an abnormal isoform (PrPsc) of the cellular prion protein (PrPc) in the brain. The scrapie prion protein, PrPsc, which is formed from PrPc, is assumed to be the etiological agent of TSE. In the transmissible particles, there is no evidence for a nucleic acid, which could be involved in the infection. Prion diseases have become an important issue not only in public health due to the possibility of plausible relationship between BSE and new variant CJD (nvCJD) but also in the scientific world due to the unique biological features of the prion agent.
    PrPc is a protein localized on the cell surface predominantly in brain, while PrPsc accumulates intracellularly in cytoplasmic vesicles. In contrast to PrPc, PrPsc has a high β-sheet content. Therefore, it is thought that this
    deleterious protein is formed by conversion of the putative α-helix of PrPc into β-sheets. No amino acid sequence or posttranslational differences have been detected between the normal host cell surface protein, PrPC, and its pathological form, PrPSc. The conversion of PrPC into PrPSc involves a conformational change whereby the α-helical content diminishes and the amount of β-sheet increases. The recognizable change between PrPC into PrPSc is acquisition of PI-PLC resistance, detergent insolubility and protease resistance. It has not been known how accumulation of PrPsc results in the subsequent changes, but there are evidences that it involves neuronal receptors, intracellular Ca2+ level and the balance of production of reactive oxygen species (ROS) and antioxidant defense mechanisms, and so on. Besides, apoptosis plays an important role in the etiology of prion disease. Apoptosis or programmed cell death is an important physiological process resulting in the controlled elimination of cells, e.g. during the development of the nervous system. In apoptosis nuclear chromatin undergoes condensation during which a calcium-dependent endonuclease is activated which cleaves nuclear DNA at linker regions, resulting in DNA fragments of 180-200bp in length and multiples of them. Apoptosis can be induced by a variety of stimuli, including heat shock, DNA damaging reagents and toxins, etc. Series of protease activation cascades involve in the apoptosis. Neuronal loss resulted from apoptosis is consistent with the absence of inflammatory react. PrP106-126, a synthetic peptide consisting of amino-acid residues 106-126 of human PrPc, is able to polymerize into amyloid-like fibrils in vitro, like PrPsc. It is neurotoxic and induces activation of astroglial and microglial cells in vitro. So PrP106-126 can serve as a model to study the cellular effects of PrPsc.
    In this experiment, the PC12 cells were infected by prion protein 106-126 peptide after differentiated by nerve growth factor (NGF). Cell viability or cell death was determined and cytotoxic effects and morphological changes were observed. The oxidative stress, energy metabolise and the calcium ion level were also detected. And the expression of Bcl-2 and Bax were detected by immunohistochemical technique. Flow cytometry and DNA electrophoresis were used to investigate whether apoptosis played a role in the pathogenesis. The activity of Caspase-3 was detected by western blot to study the function mode of apoptosis in the prion disease. The objective of this study is to evaluate the cytotoxic effects of prion protein 106-126 peptide, evaluate the possibility of the prion disease model which use differentiated PC12 cell infected by prion protein 106-126 peptide, and also evaluate the mechanism and effects of apoptosis in this model. Infected by this peptide, cell viability decreased along with the time the peptide infect and along with the dosage the peptide infect. Decrease of mitochondrial membrane potential were detected by flow cytometry, the activity of Ca2+ATPase decreased and increased reactive oxygen species (ROS) were detected by spectrophotometer ,and also the increased calcium ion in the cells were detected by confocal laser scanning microscopy. Under light microscope, cells were shrinked and rounded, many cells were divorced from plate wall, some neuraxon shortened and broken. Apoptosis cells which nucleolus shrinked and rounded could be coloured orange by fluorescent colouration. Under electron microscope, chromatin gathered along the inside of the nuclear membrane, vacuole bodies appeared. In the histogram of fluorescent staining of PI, a characteristic sub-G1 peak appeared before the diploid peak (G1/G0 phase) when treated with PrP106-126, apoptosis accounted for 28.2%, while the control group did not appear these sub-G1 peak. And also the ladder band appeared in DNA electrophoresis in the treat group. This phenomenon suggests apoptosis occurs in primary neuron culture, PrP106-126 induces neuronal apoptosis greatly. This might be a perfect model to study the cellular toxicity of prion protein.
引文
1. Jeffrey M, Gonzalez L. Pathology and pathogenesis of bovine spongiform encephalopathy and scrapie. Curr Top Microbiol Immunol. 2004, 284:65-97.
    2. Rogivue C, Gottwein J, Aguzzi A, Glatzel M. Transmissible spongiform encephalopathies in humans Schweiz Arch Tierheilkd. 2002, 144(12):685-90.
    3. Grigor'ev VB. Prion diseases of man and animal,Vopr Virusol. 2004, 49(5):4-12.
    4. Prusiner SB. Novel proteinaceous infectious particles cause scrapie. Science. 1982, 216(4542):136-44.
    5. Kitamoto T and Tateishi J. Human prion disease and human prion protein disease. Curr Top Microbiol Immunol, 1996,207: 27-34.
    6. Alter M. How is Creutzfeldt-Jakob disease acquired? Neuroepidemiology. 2000, 19(2):55-61.
    7. Collinge J. Prion diseases of humans and animals: their causes and molecular basis. Annu Rev Neurosci. 2001, 24:519-50.
    8. Priola SA, Vorberg I. Molecular aspects of disease pathogenesis in the transmissible spongiform encephalopathies. Methods Mol Biol. 2004, 268:517-40.
    9. Doerr-Schott J, Kitamoto T, Tateishi J, et al. Immunogold light and electron microscopic detection of amyloid plaques in transmissible spongiform encephalopathies. Neuropathol Appl Neurobiol. 1990, 16(1): 85-9
    10. Zeidler M, Ironside JW. The new variant of Creutzfeldt-Jakob disease. Rev Sci Tech. 2000, 19(1):98-120.
    11. Berr C, Helbecque N, Sazdovitch V, et al. Polymorphism of the codon 129 of the prion protein (PrP) gene and neuropathology of cerebral ageing. Acta Neuropathol (Berl). 2003, 106(1):71-4.
    12. Beaudry P, Cohen P, Brandel JP, et al. 14-3-3 protein, neuron-specific enolase, and S-100 protein in cerebrospinal fluid of patients with Creutzfeldt-Jakob disease. Dement Geriatr Cogn Disord. 1999, 10(1):40-6
    13. Cambier DM, Kantarci K, Worrell GA, et al. Lateralized and focal clinical, EEG, and FLAIR MRI abnormalities in Creutzfeldt-Jakob disease. Clin Neurophysiol. 2003, 114(9):1724-8.
    14. Haik S, Dormont D, Faucheux BA, et al. Prion protein deposits match magnetic resonance imaging signal abnormalities in Creutzfeldt-Jakob disease. Ann Neurol. 2002, 51(6):797-9.
    15. Montrasio F, Cozzio A, Flechsig E, et al. B lymphocyte-restricted expression of prion protein does not enable prion replication in prion protein knockout mice. Proc Natl Acad Sci U S A. 2001, 98(7):4034-7.
    16. DeArmond S J. Discovering the mechanisms of neurodegeneration in prion diseases. Neurochem Res. 2004, 29(11):1979-98.
    17. Bratosiewicz W J, Wasik TJ, Liberski PP. Molecular approaches to mechanisms of prion diseases. Folia Neuropathol. 2004, 42:33-46.
    18. Bendheim P E, H R Brown, R D Rudelli, et al. Nearly ubiquitous tissue distribution of the scrapie agent precursor protein. Neurology, 1992, 42:149-156
    19. Holada K, Simak J, Risitano A M, et al. Activated platelets of patients with paroxysmal nocturnal hemoglobinuria express cellular prion protein. Blood, 2002, 100: 341-343
    20. Harris D A, P Lele, Snider W D. Localization of the mRNA for a chicken prion protein by in situ hybridization. Proc. Natl. Acad. Sci. USA , 1993, 90:4309-4313
    21. Herms J W, H A Kretzschmar, Titz S, et al. Patch-clamp analysis of synaptic transmission to cerebellar Purkinje cells of prion protein knockout mice. Eur J Neurosci. 1995. 7:2508-2512
    22 Brown DR, Schulz-Schaeffer WJ, Schmidt B, et al. Prion protein-deficient cells show altered response to oxidative stress due to decreased SOD-1 activity. Exp Neurol, 1997, 146(1): 104-12.
    23 Tobler I, Gaus S E, Deboer T, et al. Altered circadian activity rhythms and sleep in mice devoid of prion protein. Nature, 1996. 380:639-642
    24 Stewart R S, Drisaldi B, Harris D A. A transmembrane form of the prion protein contains an uncleaved signal peptide and is retained in the endoplasmic Reticulum. Mol. Biol. Cell, 2001, 12: 881-889
    25 Pan T, Li R, Wong B-S, et al. Heterogeneity of normal prion protein in two-dimensional immunoblot: presence of various glycosylated and truncated forms. J Neurochem, 2002, 81: 1092-1101
    26 Low M G. The glycosyl-phosphatidyl anchor of membrane proteins. Acta, 1989, 988:427-454
    27 Borchelt D R, M Rogers, N Stahl, et al. Release of the cellular prion protein from cultured cells after loss of its glycoinositol phospholipid anchor. Glycobiology, 1993, 3:319-329
    28 Harris D A, A Gorodinsky, S Lehmann, et al. Cell biology of the prion protein. Curr. Top. Microbiol. Immunol. 1996, 207:77-93
    29 Quaglio E, Chiesa R, Harris D A. Copper converts the cellular prion protein into a protease-resistant species that is distinct from the scrapie isoform. J. Biol. Chem. 2001, 276: 11432-11438
    30 Caughey B, K Neary, R Buller, et al. Normal and scrapie-associated forms of prion protein differ in their sensitivities to phospholipase and proteases in intact neuroblastoma cells. J. Virol. 1990, 64:1093-1101
    31 Borchelt D R, V E Koliatsos, M Guarnieri, et al. Rapid anterograde axonal transport of the cellular prion glycoprotein in the peripheral and central nervous systems. J. Biol. Chem. 1994, 269:14711-14714
    32 Shyng S L, M T Huber, D A Harris. A prion protein cycles between the cell surface and an endocytic compartment in cultured neuroblastoma cells. J. Biol. Chem. 1993, 268:15922-15928
    33 Schatzl H M, L Laszlo, D M Holtzman, et al. A hypothalamic neuronal cell line persistently infected with scrapie prions exhibits apoptosis. J. Virol. 1997, 71:8821-8831
    34 Daude N, S Lehmann, D A Harris. Identification of intermediate steps in the conversion of a mutant prion protein to a scrapie-like form in cultured cells. J. Biol. Chem. 1997, 272:11604-11612
    35 Sabuncu E, Petit S, Le Dur A, et al. PrP polymorphisms tightly control sheep prion replication in cultured cellsJ. Virol. 2003, 77(4): 2696-2700
    36 McKinley M P, A Taraboulos, L Kenaga, et al. Ultrastructural localization of scrapie prion proteins in cytoplasmic vesicles of infected cultured cells. Lab. Investig. 1991, 65:622-630
    37 Stahl N, M A Baldwin, A L Burlingame, et al. Identification of glycoinositol phospholipid linked and truncated forms of the scrapie prion protein.
    Biochemistry 1990, 29:8879-8884
    38 Borchelt D R, A Taraboulos, S B Prusiner. Evidence for synthesis of scrapie prion proteins in the endocytic pathway. J. Biol. Chem. 1992, 267:16188-16199
    39 Daude N, S Lehmann, D A Harris. Identification of intermediate steps in the conversion of a mutant prion protein to a scrapie-like form in cultured cells. J. Biol. Chem. 1997, 272:11604-11612
    40 Zou W-Q, Cashman N R. Acidic pH and detergents enhance in vitro conversion of human brain PrPC to a PrPSc-like form. J. Biol. Chem. 2002, 277: 43942-43947
    41 Caughey B, G J Raymond, D Ernst, et al. N-terminal truncation of the scrapie-associated form of PrP by lysosomal protease(s): implications regarding the site of conversion of PrP to the protease-resistant state. J. Virol. 1991, 65:6597-6603
    42 Chen S G, D B Teplow, P Parchi, et al. Truncated forms of the human prion protein in normal brain and in prion diseases. J. Biol. Chem. 1995, 270:19173-19180
    43 Caughey B W, A Dong , K S Bhat, et al. Secondary structure analysis of the scrapie-associated protein PrP 27-30 in water by infrared spectroscopy. Biochemistry 1991, 30:7672-7680
    44 Pan K-M, M Baldwin, J Nguyen, et al. Conversion of α-helices into β-sheets features in the formation of the scrapie prion proteins. Proc. Natl. Acad. Sci. USA 1993, 90:10962-10966
    45 Come J H, P E Fraser, P T Lansbury. A kinetic model for amyloid formation in the prion diseases: importance of seeding. Proc. Natl. Acad. Sci. USA 1993, 90:5959-5963
    46 Ivanova L, Barmada S, Kummer T, et al. Mutant prion proteins are partially retained in the endoplasmic reticulum. J. Biol. Chem. 2001, 276: 42409-42421
    47 Castilla J, Hetz C, Soto C. Molecular mechanisms of neurotoxicity of pathological prion protein. Curr Mol Med. 2004, 4(4):397-403
    48 Hartl F U. Molecular chaperones in cellular protein folding. Nature. 1996, 381:571-580
    49 Welch W J, and P Gambetti. Chaperoning brain diseases. Nature. 1998, (6671) 392:23-24
    50 Kaneko K, L Zulianello, M Scott, et al. Evidence for protein X binding to a discontinuous epitope on the cellular prion protein during scrapie prion propagation. Proc. Natl. Acad. Sci. USA ,1997, 94:10069-10074
    51. J I Kourie and A A Shorthouse. Properties of cytotoxic peptide-formed ion channels. Am J Physiol Cell Physiol 2000,278: C1063-C1087
    52. Kourie J I, Culverson A L, Farrelly P V, et. al. Heterogeneous amyloid-formed ion channels as a common cytotoxic mechanism: implications for therapeutic strategies against amyloidosis. Cell Biochem Biophys. 2002, 36(2-3):191-207.
    53. Kourie J I, Henry C L. Ion channel formation and membrane-linked pathologies of misfolded hydrophobic proteins: the role of dangerous unchaperoned molecules. Clin Exp Pharmacol Physiol. 2002, 29(9):741-53.
    54. Lin M C, Mirzabekov T, Kagan B L. Channel formation by a neurotoxic prion protein fragment. J Biol Chem. 1997, 272(1):44-7.
    55 Ettaiche M, Pichot R, Vincent J P, et al. In vivo cytotoxicity of the prion protein fragment 106-126. J Biol Chem. 2000, 24;275(47):36487-90.
    56. Bounias M, Purdey M. Transmissible spongiform encephalopathies: a family of etiologically complex diseases--a review. Sci Total Environ. 2002, 297(1-3):1-19.
    57. De Gioia L, Selvaggini C, Ghibaudi E, et al. Conformational polymorphism of the amyloidogenic and neurotoxic peptide homologous to residues 106-126 of the prion protein. J Biol Chem. 1994, 269(11):7859-62.
    58. Muller W E, Ushijima H, Schroder H C, et al. Cytoprotective effect of NMDA receptor antagonists on prion protein (PrionSc)-induced toxicity in rat cortical cell cultures. Eur J Pharmacol. 1993, 246(3):261-7.
    59. Satheeshkumar K S, Murali J, Jayakumar R. Assemblages of prion fragments: novel model systems for understanding amyloid toxicity. J Struct Biol. 2004, 148(2):176-93.
    60. Singh N, Gu Y, Bose S, et al. Prion peptide 106-126 as a model for prion replication and neurotoxicity. Front Biosci. 2002, 7: 60-71.
    61. Forloni G. Neurotoxicity of beta-amyloid and prion peptides. Curr Opin Neurol. 1996 , 9(6):492-500.
    62. Tagliavini F, Forloni G, Colombo L, et al. Tetracycline affects abnormal properties of synthetic PrP peptides and PrP(Sc) in vitro. J Mol Biol. 2000, 300(5):1309-22.
    63. Salmona M, Forloni G, Diomede L, et al. A neurotoxic and gliotrophic fragment of the prion protein increases plasma membrane microviscosity. Neurobiol Dis. 1997, 4(1):47-57.
    64. Lin M C, Mirzabekov T, Kagan B L. Channel formation by a neurotoxic prion protein fragment. J Biol Chem. 1997, 3;272(1):44-7.
    65. Kourie J I, Farrelly P V, Henry C L. Channel activity of deamidated isoforms of prion protein fragment 106-126 in planar lipid bilayers. J Neurosci Res. 2001, 66(2):214-20.
    66. Brown D R, Schmidt B, Kretzschmar H A. A neurotoxic prion protein fragment enhances proliferation of microglia but not astrocytes in culture. Glia. 1996,18(1):59-67.
    67. Jesionek-Kupnicka D, Buczynski J, Kordek R, et al. Programmed cell death (apoptosis) in Alzheimer's disease and Creutzfeldt-Jakob disease. Folia Neuropathol. 1997, 35(4):233-5.
    68. Kawashima T, Doh-ura K, Ogomori K, et al. Apoptotic bodies in the cerebellum of Japanese patients with Creutzfeldt-Jakob disease. Pathol Int. 2001, 51(3):140-4.
    69. Thellung S, Florio T, Corsaro A, et al. Intracellular mechanisms mediating the neuronal death and astrogliosis induced by the prion protein fragment 106-126. Int J Dev Neuroscience, 2000, 18(4-5): 481-492.
    70. Andreyev A Y, Kushnareva Y E, Starkov A A. Mitochondrial metabolism of reactive oxygen species. Biochemistry . 2005, 70(2):200-14.
    71. Choi S I, Ju W K, Choi E K, et al. Mitochondrial dysfunction induced by oxidative stress in the brains of hamsters infected with the 263 K scrapie agent. Acta Neuropathol (Berl). 1998, 96(3):279-86.
    72. Guentchev M, Siedlak S L, Jarius C, et al. Oxidative damage to nucleic acids in human prion disease. Neurobiol Dis. 2002, 9(3):275-81.
    73. Singh K K. Mitochondria damage checkpoint in apoptosis and genome stability. FEMS Yeast Res. 2004, 5(2):127-32.
    74. Aiken J M, Williamson J L, Marsh R F. Evidence of mitochondrial involvement in scrapie infection. J Virol. 1989, 63(4):1686-94.
    75. Kim B H, Lee H G, Choi J K, et al. The cellular prion protein (PrPC) prevents apoptotic neuronal cell death and mitochondrial dysfunction induced by serum deprivation. Brain Res Mol Brain Res. 2004, 124(1):40-50.
    76. Hachiya N S, Yamada M, Watanabe K, et al. Mitochondrial localization of cellular prion protein (PrPC) invokes neuronal apoptosis in aged transgenic mice overexpressing PrPC. Neurosci Lett. 2005, 10;374(2):98-103.
    77. Silei V, Fabrizi C, Venturini G, et al. Measurement of intracellular calcium levels by the fluorescent Ca(2+) indicator Calcium-Green. Brain Res Brain Res Protoc. 2000, 5(2):132-4.
    78. Sandberg M K, Wallen P, Wikstrom M A, et al. Scrapie-infected GT1-1 cells show impaired function of voltage-gated N-type calcium channels (Ca(v) 2.2) which is ameliorated by quinacrine treatment. Neurobiol Dis. 2004 ,15(1):143-51.
    79. Thellung S, Florio T, Villa V, et al. Apoptotic cell death and impairment of L-type voltage-sensitive calcium channel activity in rat cerebellar granule cells treated with the prion protein fragment 106-126. Neurobiol Dis. 2000, 7(4):299-309.
    80. Offen D, Elkon H, Melamed E. Apoptosis as a general cell death pathway in neurodegenerative diseases. J Neural Transm Suppl. 2000, (58):153-66.
    81. Lucassen PJ, Williams A, Chung W C, et al. Detection of apoptosis in murine scrapie. Neurosci Lett, 1995, 198(3): 185-188.
    82. Gray F, Chretien F, Adle-Biassette H, et al. Neuronal apoptosis in Creutzfeldt-Jakob disease. J Neuropathol Exp Neurol, 1999, 58(4): 321-328.
    83. Forloni G, Bugiani O, Tagliavini F, et,al. Apoptosis-mediated neurotoxicity induced by beta-amyloid and PrP fragments. Mol Chem Neuropathol. 1996, 28(1-3):163-71.
    84. Forloni G, Angeretti N, Chiesa R, et al. Neurotoxicity of a prion protein fragment. Nature. 1993, 8;362(6420):543-6.
    85. Florio T, Thellung S, Amico C,et al. Prion protein fragment 106-126 induces apoptotic cell death and impairment of L-type voltage-sensitive calcium channel activity in the GH3 cell line. J Neurosci Res. 1998, 54(3):341-52.
    86. Heiser D, Labi V, Erlacher M, et al. The Bcl-2 protein family and its role in the development of neoplastic disease. Exp Gerontol. 2004, 39(8):1125-35.
    87. Sorenson C M. Bcl-2 family members and disease. Biochim Biophys Acta. 2004, 1644(2-3):169-77.
    88. Ookohchi T, Ito H, Serikawa T, et al. Detection of apoptosis in the brain of the zitter rat with genetic spongiform encephalopathy. Biochem Mol Biol Int. 1997, 41(2):279-84.
    89. Perovic S, Schroder H C, Pergande G, et al. Effect of flupirtine on Bcl-2 and glutathione level in neuronal cells treated in vitro with the prion protein fragment (PrP106-126). Exp Neurol. 1997, 147(2):518-24.
    90. Hetz C, Russelakis-Carneiro M, Maundrell K, et al. Caspase-12 and endoplasmic reticulum stress mediate neurotoxicity of pathological prion protein. EMBO J. 2003, 22(20):5435-45.
    91. Kurschner C, Morgan J I. The cellular prion protein (PrP) selectively binds to Bcl-2 in the yeast two-hybrid system. Brain Res Mol Brain Res. 1995,
    30(1):165-8.
    92. Sakudo A, Lee DC, Saeki K, et al. Tumor necrosis factor attenuates prion protein-deficient neuronal cell death by increases in anti-apoptotic Bcl-2 family proteins. Biochem Biophys Res Commun. 2003, 310(3):725-9.
    93. Siso S, Puig B, Varea R, et al. Abnormal synaptic protein expression and cell death in murine scrapie. Acta Neuropathol (Berl). 2002, 103(6):615-26.
    94. Bounhar Y, Zhang Y, Goodyer CG, et al. Prion protein protects human neurons against Bax-mediated apoptosis. J Biol Chem. 2001, 19;276(42):39145-9.
    95. Erwan Paitel, Robin Fahraeus, and Frédéric Checler. Cellular Prion Protein Sensitizes Neurons to Apoptotic Stimuli through Mdm2-regulated and p53-dependent Caspase 3-like Activation. J. Biol. Chem. 2003,278: 10061 -10066.
    96. Chiesa R, Piccardo P, Dossena S, et al. Bax deletion prevents neuronal loss but not neurological symptoms in a transgenic model of inherited prion disease. Proc Natl Acad Sci U S A. 2005,102(1):238-43.
    97. Gibson B W. The human mitochondrial proteome: oxidative stress, protein modifications and oxidative phosphorylation. Int J Biochem Cell Biol. 2005, 37(5):927-34.
    98. Saez-Valero J, Angeretti N, Forloni G. Caspase-3 activation by beta-amyloid and prion protein peptides is independent from their neurotoxic effect. Neurosci Lett. 2000, 3;293(3):207-10.
    99. Paitel E, Alves da Costa C, Vilette D, et al. Overexpression of PrPc triggers caspase 3 activation: potentiation by proteasome inhibitors and blockade by anti-PrP antibodies. J Neurochem. 2002, 83(5):1208-14.
    100. Stennicke H R. Caspases--at the cutting edge of cell death. Symp Soc Exp Biol. 2000, 52:13-29.
    101. Rubenstein R, Carp R I, Callahan S M. In vitro replication of scrapie agent in a neuronal model: infection of PC12 cells. J Gen Virol. 1984, 65 ( Pt 12):2191-8.
    102. Rubenstein R, Deng H, Scalici C L, et al. Alterations in neurotransmitter-related enzyme activity in scrapie-infected PC12 cells. J Gen Virol. 1991, ( 6):1279-85.
    103. Chandler R L. Encephalopathy in mice produced by inoculation with scrapie brain material. Lancet, 1961, 1: 1378-1379.
    104. Gajdusek D C, Gibbs C J, Alpers M, et al. Experimental transmission of a Kuru-like syndrome to chimpanzees. Nature, 1966, 209(25): 794-796.
    105. Tateishi J, Kitamoto T, Hoque MZ, et al. Experimental transmission of Creutzfeldt-Jakob disease and related diseases to rodents. Neurology, 1996, 46(2): 532-537.
    106. Jeffrey M, Scott JR, Williams A, et al. Ultrastructural features of spongiform encephalopathy transmitted to mice from three species of bovidae. Acta Neuropathol(Berl), 1992, 84(5): 559-569.
    107. Pattison I H, Jones K M. Modification of a strain of mouse-adapted scrapie by passage through rats. Res Vet Sci, 1968, 9(5): 408-410.
    108. Mallucci G R, Ratte S, Asante E A, et al. Post-natal knockout of prion protein alters hippocampal CA1 properties, but does not result in neurodegeneration.
NGLC 2004-2010.National Geological Library of China All Rights Reserved.
Add:29 Xueyuan Rd,Haidian District,Beijing,PRC. Mail Add: 8324 mailbox 100083
For exchange or info please contact us via email.