促红细胞生成素对帕金森病模型的保护作用及其机制的实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
第一部分促红细胞生成素受体在1-甲基-4-苯基-1, 2, 3, 6-四氢吡啶致C57BL/6小鼠多巴胺能神经元损伤模型不同脑区的表达变化
     目的探讨促红细胞生成素受体(Erythropoietin receptor, EPOR)在1-甲基-4-苯基-1,2,3,6-四氢吡啶(1-methy1-4-phenvl-1, 2, 3, 6-tetrahvdropvridine, MPTP)致C57BL/6小鼠多巴胺能神经元损伤模型中不同脑区不同时程的表达变化。
     方法采用腹腔注射MPTP制备C57BL/6小鼠PD模型。36只雄性C57BL/6小鼠,随机分为7组(n=12只):对照组、MPTP1d组、MPTP2d组、MPTP4d组、MPTP7d组、MPTP14d组、MPTP21d组。在末次注射MPTP后的相应时间将动物处死,Western blot法检测腹侧中脑、纹状体、皮质区EPOR的表达;免疫组化法检测腹侧中脑、纹状体、皮质区EPOR阳性细胞数。
     结果MPTP致伤显著增加C56BL/6小鼠腹侧中脑EPOR的表达,在末次注射MPTP4天后开始升高,并且进行性增加至21天。MPTP对纹状体区与皮质区EPOR的表达均无显著性影响。
     结论MPTP所致腹侧中脑区EPOR蛋白表达增加可能与PD发生发展的病理机制有关。
     第二部分促红细胞生成素对1-甲基-4-苯基吡啶离子致PC12细胞损伤的抗凋亡作用及其机制研究
     目的探讨促红细胞生成素(Erythropoietin,EPO)对1-甲基-4-苯基吡啶离子(1-methy14-phenylpyridinium,MPP+)诱导PC12细胞变性损伤的保护作用及其机制。方法以MPP+损伤PC12细胞为帕金森病细胞模型,将培养的PC12细胞,分为3组:空白对照组、MPP+组、EPO+MPP+组。对照组给予无血清的DMEM培养基培养,MPP+组给予500μmol/L的MPP+处理24 h,EPO+MPP+组给予不同浓度的EPO(0.1U、0.3 U、1 U、3 U、10 U/ml)与500μmol/L的MPP+同时处理24 h。上述各组细胞加药孵育24 h后,采用四甲基偶氮唑盐(MTT)法检测细胞存活率,流式细胞术与TUNEL法检测细胞凋亡,2′,7′-dichlorofluorescin diacetate (DCF-DA)染色检测细胞内活性氧(ROS)的含量,罗丹明123 (Rhodamine123, Rh123)染色检测细胞线粒体膜电位(△Ψm),荧光法检测Caspase-3活性,Western blot法检测Bax与Bcl-2蛋白表达。
     结果①500μmol/L的MPP+可以导致PC12细胞存活率下降,凋亡率增加;0.1-10 U/ml的EPO可以使PC12细胞存活率增加,凋亡率降低,并在1U/ml时达到最大保护效应;②MPP+导致ROS含量增加、△Ψm降低、Caspase-3活性增加; EPO可以抑制MPP+所致ROS、△Ψm和Caspase-3水平的变化;③MPP+组较空白对照组相比,Bax/Bcl-2比率增加;而EPO-MPP+组较MPP+组相比,Bax/Bcl-2比率降低。
     结论EPO对MPP+致PC12细胞损伤具有保护作用,其机制是通过EPO抗氧化应激及抗凋亡的特性而实现的。
     第三部分促红细胞生成素对MPP+致PC12细胞损伤的保护作用通路
     实验一Akt/GSK-3β/Caspase-3通路在促红细胞生成素对MPP+致PC12细胞损伤保护中的作用
     目的研究EPO对MPP+致PC12细胞损伤保护作用中Akt/GSK-3β/Caspase-3通路的作用。
     方法以500μmol/L的MPP+损伤PC12细胞为帕金森病细胞模型,实验分组如下:空白对照组、LY294002组、LiCl组、MPP+组、MPP++EPO组、MPP++LiCl组、MPP++EPO+LY294002组。空白对照组、LY294002组、LiCl组、MPP+组、MPP++EPO组分别给予无血清DMEM培养基、10μmol/L的LY294002、20 mmol/L的LiCl、500μmol/L的MPP+、500μmol/L的MPP+ +1 U/ml的EPO处理;MPP++LiCl组先给予20 mmol/L的LiCl,1 h后给予500μmol/L的MPP+处理;MPP++EPO+LY294002组先给予10μmol/L的LY294002,1 h后给予500μmol/L的MPP+ +1 U/ml的EPO处理。孵育24 h后采用MTT法检测细胞存活率,TUNEL法检测细胞凋亡;孵育0.5 h、1 h、3 h、6 h、12 h之后采用检测Western免疫印迹法检测Akt、p-Akt、GSK-3β、p-GSK-3β蛋白表达;孵育0 h、4 h、8 h、16 h之后采用荧光法检测Caspase-3活性的变化。
     结果①MPP+和/或EPO均可以诱导p-Akt表达增加;加用LY294002后p-Akt表达降低。②应用LY294002拮抗了EPO对于PC12细胞的保护作用。③MPP+和/或EPO均可以诱导p-GSK-3β表达增加;加用LY294002后p-GSK-3β表达降低。④应用LiCl拮抗了MPP+对于PC12细胞的损伤作用。⑤MPP+致PC12细胞内Caspase-3活性增加,EPO和LiCl对MPP+所致的Caspase-3活性增加均有抑制作用;应用LY294002拮抗了EPO对Caspase-3活性的抑制作用。
     结论EPO对MPP+致PC12细胞损伤的保护作用是通过Akt/GSK-3b/Caspase-3通路发挥作用。.
     实验二ERK通路在促红细胞生成素对MPP+致PC12细胞损伤保护中的作用
     目的研究EPO对MPP+致PC12细胞损伤保护作用中ERK通路的作用。
     方法以500μmol/L的MPP+损伤PC12细胞为帕金森病细胞模型,实验分组如下:空白对照组、PD98059组、MPP+组、MPP++EPO组、MPP++EPO+PD98059组。空白对照组、PD98059组、MPP+组、MPP++EPO组分别给予无血清DMEM培养基、50μmol/L的PD98059、500μmol/L的MPP+、500μmol/L的MPP+ +1 U/ml的EPO处理; MPP++EPO+ PD98059组先给予50μmol/L的PD98059 ,1 h后给予500μmol/L的MPP+ +1 U/ml的EPO处理。上述各组孵育24 h后采用四甲基偶氮唑盐(MTT)法检测细胞存活率,TUNEL法检测细胞凋亡;孵育0.5 h、1 h、3 h、6 h、12 h之后采用Western免疫印迹法检测p-ERK、ERK蛋白表达。
     结果①MPP+和/或EPO均可以诱导p-ERK表达短暂增加;加用PD98059后p-ERK表达降低。②应用PD98059并不能拮抗EPO对于PC12细胞的保护作用。
     结论EPO可以短暂激活ERK通路,但ERK通路并没有在EPO对MPP+致PC12细胞损伤的保护作用中发挥作用。
Part I The change of expression of erythropoietin receptor in certain brain regions of 1-methy1-4-phenvl-1, 2, 3, 6-tetrahvdropvridine Parkinson disease model mices
     Objective To investigate the change of expression of erythropoietin receptor (EPOR) in certain brain regions on 1-methy1-4-phenvl-1, 2, 3, 6-tetrahvdropvridine (MPTP) lesioned Parkison disease model mice.
     Methods①Establishing MPTP lesioning of PD model mice: At the beginning of the experiment, the mice (12 per group) received an intraperitoneal (i.p.) injection of MPTP-HCl in saline at 24 h intervals for 5 days. The control group was injected with saline only. The mice were sacrificed at 1, 2, 4, 7, 14 and 21 days after the last MPTP injection.②Western blot: Western blot was carried out using total protein isolated from ventral mesencephalon, striatum and cortex, which were removed rapidly on ice from decapitated mice.③Immunohistochemistry: Immunohistochemistry experiments were carried out using tissue section prepared from ventral mesencephalon, striatum and cortex, which rapidly removed from sacrificed mice. Change of the number of EPO-R positive cells was identified using hemi-quantitive method.
     Results 4 day after the last MTPT treatment, the expression of EPOR and the number of EPO-R positive cells significantly increased in ventral mesencephalon compared to control group, the increased tendency continued until the 21day. While these index in striatum and cortex were not statistically different compared with those in animals that did not undergo MPTP insult.
     Conclusion The results showed that EPOR expression in ventral mesencephalon increased significantly after MPTP injection which strongly indicated that there were certain relationships between EPOR and the development of PD.
     Part II Protective effects and mechanisms of erythropoietin on 1-methy14-phenylpyridinium -induced neurodegeneration in PC12 cells
     Objective The neuroprotective effect of erythropoietin (EPO) against 1-methyl-4- phenylpyridinium (MPP+)-induced oxidative stress in cultured PC12 cells, as well as the underlying mechanism, were investigated.
     Methods PC12 cells impaired by MPP+ were used as the cell model of Parkinson’s disease. The cultured cells were divide into 3 groups: control group was treated with DMEM medium, MPP+ group was treated with 500μmol/L MPP+, MPP+ and EPO group was co-incubated with 500μmol/L MPP+ and different concentrations (0, 0.1, 0.3, 1, 3, or 10 U/mL) of EPO. After incubation for 24 h, methyl thiazolyl tetrazolium (MTT) was used to assay the viability of the PC12 cells exposed to gradient concentrations of EPO, flow cytometry and the terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL) assay were used to analyze the apoptosis ratio of PC12 cells. The expression of Bcl-2 and Bax in PC12 cells were examined by Western blot, and the reactive oxygen species (ROS), the mitochondrial transmembrane potential and the activity of Caspase-3 in each group were detected by spectrofluorometer.
     Results①Treatment of PC12 cells with MPP+ caused the loss of cell viability, which may be associated with the elevation in apoptotic rate, EPO had significantly protective effect on the neurodegeneration and had the maximum protective effect at 1 U/ml.②MPP+ induced the formation of ROS, the disruption of mitochondrial transmembrane potential and the activation of Caspase-3. In contrast, EPO significantly reversed these responses.③It was also shown that MPP+ significantly induced the upregulation of Bax/Bcl-2 ratio, while EPO significantly downregulated the ratio.
     Conclusion EPO may provide a useful therapeutic strategy for treatment of neurodegenerative diseases such as Parkinson’s disease, and the inhibitive effect of EPO on the MPP+-induced cytotoxicity may be ascribed to its anti-oxidative property and anti-apoptotic activity.
     Part III The study of signaling pathway in the protective effect of EPO on MPP+ induced neurotoxicity in PC12 cells
     Section I The role of Akt/GSK-3β/Caspase-3 signaling pathway in the protective effect of EPO on MPP+ induced neurotoxicity in PC12 cells Objective To investigate the role of Akt/GSK-3β/Caspase-3 signaling pathway in the protective effect of EPO on MPP+ induced neurotoxicity in PC12 cells.
     Method PC12 cells impaired by MPP+ were used as the cell model of Parkinson’s disease. The cultured cells were divide into such groups: control group received the administration of DMEM medium; LY294002 group, LiCl group and MPP+ group were identical to the control group except that LY294002 (10μmol/L), LiCl (20 mmol/L) or MPP+ (500μmol/L) was administered instead of DMEM medium, respectively; MPP++EPO group was treated with MPP+(500μmol/L) and EPO(1U/ml); MPP++LiCl group received LiCl (20 mmol/L) 1 h before treatment with MPP+(500μmol/L); MPP++EPO+LY294002 group was identical to the MPP++EPO group except that LY294002 was given 1 h before MPP+ and EPO. After incubation for 24 h, methyl thiazolyl tetrazolium (MTT) was used to assay the viability of the PC12 cells and the terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL) assay were used to analyze the apoptosis ratio of PC12 cells. After incubation for 0.5 h, 1 h, 3 h, 6 h, 12 h, western blot was used to detect the expression level of Akt, p-Akt, GSK-3β, p-GSK-3β. After incubation for 0 h, 4 h, 8 h, 16 h, the activity of Caspase-3 was detected by spectrofluorometer.
     Results①Enhanced levels of p-Akt were detected EPO was added to the culture and there was a significant increase in the phosphorylation of Akt in MPP+-treated cells by EPO. LY294002 thoroughly abolished EPO-induced phosphorylation of Akt.②The effect of rescuing the cell from death induced by EPO was lost with addition of LY 294002, a specific inhibitor of PI3K.③Phosphorylation of GSK-3βwas enhanced following EPO-treated and there was a significant increase in the phosphorylation of GSK-3βin MPP+-treated cells by EPO. LY294002 pretreatment abolished phosphorylation of GSK-3βby EPO.④Inhibition of GSK-3βby LiCl promoted viability and reduced apoptosis in PC12 cells subjected to MPP+.⑤MPP+ induced a time dependent increase in caspase-3-like proteinase activities. EPO and LiCl significantly decreased caspase 3-like activities induced by MPP+ toxicity. The addition of the PI3K inhibitor, LY294002 in PC12 cells reversed the effect of EPO on Caspase-3 activation.
     Conclusion Thus, these findings indicate that EPO protects against apoptosis in PC12 cells exposed to MPP+, through the Akt/GSK-3b/ Caspase-3 signaling pathway in this model system. EPO;; MPP~+ ;; PC12 cell;; ERK;; apoptosis
引文
1. Chung YH, Kim SI, Joo KM, et al. Age-related changes in erythropoietin immunoreactivity in the cerebral cortex and hippocampus of rats. Brain Res, 2004, 1018:141-146.
    2. Siren AL, Fratelli M, Brines M, et al. Erythropoietin prevents neuronal apoptosis after cerebral ischemia and in metabolic stress. Proc Natl AcadSci USA, 2001, 98:4044–4049.
    3. Ruscher K, Freyer D, Karsch M, et al. Erythropoietin is a paracrine mediator of ischemic tolerance in the brain: evidence from an in vitro model. J Neurosci, 2002, 22:10291–10301.
    4. Chong ZZ, Kang JQ, Maiese K. Erythropoietin forsters both intrinsic and extrinsic neuronal protection through modulation of microglia, Akt-a, Bad, and caspase-mediated pathways. Br J Pharmacol, 2003, 138:1107–1118.
    5. Ghezzi P, Brines M. Erythropoietin as an anti-apoptotic, tissue neuroprotective cytokine. Cell Death Diff, 2004, 11: S37–44.
    6. Csete M, Rodriguez L, Wilcox M, et al. Erythropoietin receptor is expressed on adult rat dopaminergic neurons and erythropoietin is neurotrophic in cultured dopaminergic neuroblasts. Neurosci Lett, 2004, 359: 124-126.
    7. Genc S, Kuralay F, Genc K, et al. Erythropoietin exerts neuroprotection in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated C57/BL mice via increasing nitric oxide production. Neurosci Lett, 2001, 298: 139-141.
    8. Siren AL, Fratelli M, Brines M, et al. Erythropoietin prevents neuronal apoptosis after cerebral ischemia and in metabolic stress. Proc Natl AcadSci USA, 2001, 98:4044–4049.
    9. Ruscher K, Freyer D, Karsch M, et al. Erythropoietin is a paracrine mediator of ischemic tolerance in the brain: evidence from an in vitro model. J Neurosci, 2002, 22:10291–10301.
    10. Chong ZZ, Kang JQ, Maiese K. Erythropoietin forsters both intrinsic and extrinsic neuronal protection through modulation of microglia, Akt-a, Bad, and caspase-mediated pathways. Br J Pharmacol, 2003, 138:1107–1118.
    11. Ghezzi P, Brines M. Erythropoietin as an anti-apoptotic, tissue neuroprotective cytokine. Cell Death Diff, 2004, 11:S37–44.
    1. Sire′n AL, Ehrenreich H. Erythropoietin-a novel concept for neuroprotection. Eur Arch Psychiatry Clin Neurosci, 2001, 251: 179–184.
    2. Brines M, Cerami A. Emerging biological roles for erythropoietin in the nervous system. Nat Rev Neurosci, 2005, 6: 484–494.
    3. Sire′n AL, Knerlich F, Poser W, et al. Erythropoietin and erythropoietin receptor in human ischemic/hypoxic brain. Acta Neuropathol, 2001, 101:271–276.
    4. Chin K, Yu X, Beleslin-Cokic B, et al. Production and processing of erythropoietin receptor transcripts in brain. Mol Brain Res, 2000, 81: 29–42.
    5. Bernaudin M, Bellail A, Marti HH, et al. Neurons and astrocytes express EPO mRNA: oxygen-sensing mechanisms that involve the redox-state of the brain. Glia, 2000, 30: 271–278.
    6. Sugawa M, Sakurai Y, Ishikawa-Ieda Y, et al. Effects oferythropoietin on glial cell development; oligodendrocyte maturation and astrocyte proliferation. Neurosci Res, 2002, 44:391–403.
    7. Nagai A, Nakagawa E, Choi HB, et al. Erythropoietin and erythropoietin receptors in human CNS neurons, astrocytes, microglia, and oligodendrocytes grown in culture. J Neuropathol Exp Neurol, 2001, 60: 386–392.
    8. Yamaji R, Okada T, Moriya M, et al. Brain capillary endothelial cells express two forms of erythropoietin receptor mRNA. Eur J Biochem, 1996, 239: 494–500.
    9. Csete M, Rodriguez L, Wilcox M, et al. Erythropoietin receptor is expressed on adult rat dopaminergic neurons and erythropoietin is neurotrophic in cultured dopaminergic neuroblasts. Neurosci Lett, 2004, 359: 124–126.
    10. Sayre LM. Biochemical mechanism of action of the dopaminergic neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Toxicol Lett, 1989, 48:121-149.
    11. Albrecht PJ, Dahl JP, Stoltzfus OK, et al. Ciliary neurotrophic factor activates spinal cord astrocytes, stimulating their production and release of fibroblast growth factor-2, to increase motor neuron survival. Exp Neurol, 2002, 173:46-62.
    12. Bakhiet M, Tjernlund A, Mousa A, et al. RANTES promotes growth and survival of human first-trimester forebrain astrocytes. Nat Cell Biol 2001, 3:150-157.
    13. Takuma K, Yoshida T, Lee E, et al. CV-2619 protects cultured astrocytes against reperfusion injury via nerve growth factor production. Eur J Pharmacol, 2000, 406: 333-339.
    14. Yamamuro A, Ago Y, Takuma K, et al. Possible involvement of astrocytes in neuroprotection by the cognitive enhancer T-588. Neurochem Res, 2003, 28: 1779-1783.
    15. Masuda S, Okano M, Yamagishi K, et al. A novel site of erythropoietin production. Oxygen-dependent production in cultured rat astrocytes. J Biol Chem, 1994, 269: 19488-19493.
    16. Diaz Z, Assaraf MI, Miller WH Jr, et al. Astroglial cytoprotection by erythropoietin pre-conditioning: Implications for ischemic and degenerative CNS disorders. J Neurochem, 2005, 93: 392-402.
    17. Siren AL, Fratelli M, Brines M, et al. Erythropoietin prevents neuronal apoptosis after cerebral ischemia and in metabolic stress. Proc Natl AcadSci USA, 2001, 98:4044–4049.
    18. Ruscher K, Freyer D, Karsch M, et al. Erythropoietin is a paracrine mediator of ischemic tolerance in the brain: evidence from an in vitro model. J Neurosci, 2002, 22:10291–10301.
    19. Chong ZZ, Kang JQ, Maiese K. Erythropoietin forsters both intrinsic and extrinsic neuronal protection through modulation of microglia, Akt-a, Bad, and caspase-mediated pathways. Br J Pharmacol, 2003, 138:1107–1118.
    20. Ghezzi P, Brines M. Erythropoietin as an anti-apoptotic, tissue neuroprotective cytokine. Cell Death Diff, 2004, 11:S37–44.
    21. Hayley S, Crocker SJ, Smith PD, et al. Regulation of dopaminergic loss by Fas in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of Parkinson’s disease. J Neurosci, 2004, 24: 2045–2053.
    22. Genc S, Akhisaroglu M, Kuralay F, et al. Erythropoietin restores glutathione peroxidase activity in 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine-induced neurotoxicity in C57BL mice and stimulates murine astroglial glutathione peroxidase production in vitro. Neurosci Lett, 2002, 321: 73-76.
    23. Snyder SH, D'Amato RJ. MPTP: a neurotoxin relevant to the pathophysiology of Parkinson's disease. The 1985 George C. Cotzias lecture. Neurology, 1986, 36:250-258.
    24. Juul SE, Yachnis AT, Rojiani AM, et al. Immunohistochemical localization of erythropoietin and its receptor in the developing human brain. Pediatr Dev Pathol, 1999, 2: 148–158.
    25. Lewczuk P, Hasselblatt M, Kamrowski-Kruck H, et al. Survival of hippocampal neurons in culture upon hypoxia: effect of erythropoietin. Neuroreport, 2000, 11: 3485– 3488.
    26. Brines ML, Ghezzi P, Keenan S, et al. Erythropoietin crosses the blood–brain barrier to protect against experimental brain injury. Proc Natl Acad Sci, 2000, 97: 10526– 10531.
    27. Sirén AL, Knerlich F, Poser W, et al. Erythropoietin and erythropoietin receptor in human ischemic/hypoxic brain. Acta Neuropathol, 2001, 101: 271-276.
    28. Grasso G, Sfacteria A, Passalacqua M, et al. Erythropoietin and erythropoietin receptor expression after experimental spinal cord injury encourages therapy by exogenous erythropoietin. Neurosurgery, 2005, 56:821-827.
    29. Eid T, Brines ML, Cerami A, et al. Increased expression of erythropoietin receptor on blood vessels in the human epileptogenic hippocampus with sclerosis. J Neuropathol Exp Neurol, 2004, 63: 73-83.
    30. Assaraf MI, Diaz Z, Liberman A, et al. Brain erythropoietin receptor expression in Alzheimer disease and mild cognitive impairment. J Neuropathol Exp Neurol, 2007, 66: 389-398.
    1. Lotharius J, O'Malley KL. The parkinsonism-inducing drug 1-methyl- 4-phenylpyridinium triggers intracellular dopamine oxidation. A novel mechanism of toxicity. J Biol Chem, 2000, 275: 38581–38588.
    2. Itano Y, Kitamura Y, Nomura Y. 1-Methyl-4-phenylpyridinium (MPP+)-induced cell death in PC12 cells: inhibitory effects of several drugs. Neurochem Int, 1994, 25: 419–424.
    3. Li Y, Takemura G, Okada H, et al. Reduction of inflammatory cytokine expression and oxidative damage by erythropoietin in chronic heart failure. Cardiovasc Res, 2006, 71: 684–694.
    4. Nakamura T, Sugaya T, Kawagoe Y, et al. Effect of erythropoietin on urinary liver-type fatty-acid-binding protein in patients with chronic renal failure and anemia. Am J Nephrol, 2006, 26: 276–280.
    5. Kumral A, Tugyan K, Gonen S, et al. Protective effects of erythropoietin against ethanol-induced apoptotic neurodegenaration and oxidative stress in the developing C57BL/6 mouse brain. Dev Brain Res, 2005, 160: 146–156.
    6. Ehrenreich H, Aust C, Krampe H, et al. Erythropoietin: novel approaches to neuroprotection in human brain disease. Metab Brain Dis, 2004, 19: 195–206.
    7. Yamamoto T, Yuyama K, Nakamura K, et al. Kinetic characterization of the nitric oxide toxicity for PC12 cells: effect of half-life time of NO release. Eur. J. Pharmacol, 2000, 397: 25–33.
    8. Vermes I, Haanen C, Steffens Nakken H, et al. A novel assay for apoptosis: Flow cytometric detection of phosphatidylserine expression on early apoptotic cells using fluorescein labeled annexin V. J Immunol Methods, 1995, 184: 39–51.
    9. Ohgoh M, Kimura M, Ogura H, et al. Apoptotic cell death of cultured cerebral cortical neurons induced by withdrawal of astroglial trophic support. Exp Neurol, 1998, 149: 51–63.
    10. Curtin JF, Donovan M, Cotter TG. Regulation and measurement of oxidative stress in apoptosis. J Immunol Methods, 2002, 265: 49-72.
    11. Lautraite S, Bigot Lasserre D, Bars R, et al. Optimization of cell-based assays for medium throughput screening of oxidative stress. Toxicol In Vitro, 2003, 17: 207–220.
    12. Wang H, Joseph JA. Quantifying cellular oxidative stress by dichlorofluorescein assay using microplate reader. Free Radic Biol Med, 1999, 27: 612–616.
    13. Lee CS, Han ES, Lee WB. Antioxidant effect of phenelzine on MPP+-induced cell viability loss in differentiated PC12 cells. Neurochem Res, 2003, 28: 1833–1841.
    14. Csete M, Rodriguez L, Wilcox M, et al. Erythropoietin receptor is expressed on adult rat dopaminergic neurons and erythropoietin is neurotrophic in cultured dopaminergic neuroblasts. Neurosci Lett, 2004, 359: 124–126.
    15. Siren AL, Fratelli M, Brines M, et al. Erythropoietin prevents neuronal apoptosis after cerebral ischemia and metabolic stress. Proc Natl Acad Sci USA, 2001, 98: 4044–4049.
    16. Chong ZZ, Lin SH, Kang JQ, et al. Erythropoietin prevents early and late neuronal demise through modulation of Akt1 and induction of caspase 1, 3, and 8. J Neurosci Res, 2003, 71:659–669.
    17. Cleeter MW, Cooper JM, Schapira AH. Irreversible inhibition of mitochondrial complex I by 1-methyl-4-phenylpyridinium: evidence for free radical involvement. J Neurochem, 1992, 58: 786–789.
    18. Sriram K, Pai KS, Boyd MR, et al. Evidence for generation of oxidative stress in brain by MPTP: in vitro and in vivo studies in mice. Brain Res. 1997, 749: 44–52.
    19. Cassarino DS, Parks JK, Parker Jr WD, et al. The Parkinsonian neurotoxin MPP+ opens the mitochondrial permeability transition pore and releases cytochrome c in isolated mitochondria via an oxidative mechanism. Biochim Biophys Acta, 1999, 1453: 49–62.
    20. Lotharius J, Dugan LL, O'Malley KL. Distinct mechanisms underlie neurotoxin-mediated cell death in cultured dopaminergic neurons. J Neurosci, 1999, 19: 1284–1293.
    21. Lee CS, Han ES, Jang YY, et al. Protective effect of harmalol and harmaline on MPTP neurotoxicity in the mouse and dopamine-induced damage of brain mitochondria and PC12 cells. J Neurochem, 2000, 75: 521–531.
    22. Blum D, Torch S, Lambeng N, et al. Molecular pathways involved in the neurotoxicity of 6-OHDA, dopamine and MPTP: contribution to the apoptotic theory in Parkinson's disease. Prog Neurobiol, 2001, 65: 135–172.
    23. Liu J, Narasimhan P, Song YS, et al. Epo protects SOD2-deficient mouse astrocytes from damage by oxidative stress. Glia, 2006, 53: 360–365.
    24. Ozturk E, Demirbilek S, Kadir But A, et al. Antioxidant properties of propofol and erythropoietinafter closed head injury in rats. Prog Neuro-psychopharmacol Biol Psychiatry, 2005, 29: 922–927.
    25. Genc S, Akhisaroglu M, Kuralay F, et al. Erythropoietin restores glutathione peroxidase activity in 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine-induced neurotoxicity in C57BL mice and stimulates murine astroglial glutathione peroxidase production in vitro. Neurosci. Lett, 2002, 321: 73–76.
    26. Zamzami N, Marchetti P, Castedo M, et al. Inhibitors of permeability transition interfere with the disruption of the mitochondrial transmembrane potential during apoptosis. FEBS Lett, 1996, 384: 53-57.
    27. Zamzami N, Marchetti P, Castedo M, et al. Reduction in mitochondrial potential constitutes an early irreversible step of programmed lymphocyte death i n vivo. J Exp Med, 1995, 181: 1661-1672.
    28. Cory S, Adams JM. The Bcl-2 family: regulators of cellular life-ordeath switch. Nat Rev Cancer, 2002, 2: 647–656.
    29. Kane DJ, Sarafian TA, Anton R, et al. Bcl-2 inhibition of neural death: decreased generation of reactive oxygen species. Science, 1993, 262: 1274–1277.
    30. Lud Cadet J, Harrington B, Ordonez S. Bcl-2 overexpression attenuates dopamine-induced apoptosis in an immortalized neural cell line by suppressing the production of reactive oxygen species. Synapse, 2000, 35: 228–233.
    31. Kirkland RA, Windelborn JA, Kasprzak JM, et al. A Baxinduced pro-oxidant state is critical for cytochrome c release during programmed neuronal death. J Neurosci, 2002, 22: 6480–6490.
    32. Renzi MJ, Farrell FX, Bittner A, et al. Erythropoietin induces changes in gene expression in PC-12 cells. Mol Brain Res, 2002, 104: 86–95.
    33. Wen TC, Sadamoto Y, Tanaka J, et al. Erythropoietin protects neurons against chemical hypoxia and cerebral ischemic injury by up-regulating Bcl-xL expression. J Neurosci Res, 2002, 67: 795–803.
    34. Chong ZZ, Kang JQ, Maiese K. Apaf-1, Bcl-xL, cytochrome c, and caspase-9 form the critical elements for cerebral vascular protection by erythropoietin. J Cereb Blood Flow Metab, 2003, 23: 320–330.
    35. Maiese K, Li F, Chong ZZ. New avenues of exploration for erythropoietin. JAMA, 2005, 293: 90–95.
    36. O'Malley, KL, Liu J, Lotharius J, et al. Targeted expression of Bcl-2 attenuates MPP+ but not 6-OHDA induced cell death in dopaminergic neurons. Neurobiol Dis, 2003, 14: 43–51.
    37. Genc S, Kuralay F, Genc K, et al. Erythropoietin exerts neuroprotection in 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine-treated C57/BL mice via increasing nitric oxide production. Neurosci Lett, 2001, 298: 139–141.
    38. Signore AP, Weng Z, Hastings T, et al. Erythropoietin protects against 6-hydroxydopamine-induced dopaminergic cell death. J Neurochem, 2006, 96: 428-443.
    1. Marti HH, Wenger RH, Rivas LA, et al. Erythropoietin gene expression in human monkey and murine brain. Eur J Neurosci, 1996, 8: 666–676.
    2. Morishita E, Masuda S, Nagao M, et al. Erythropoietin receptor is expressed in rat hippocampal and cerebral cortical neurons, and erythropoietin prevents in vitro glutamate induced neuronal death. Neuroscience, 1997, 76: 105–116.
    3. Chong ZZ, Lin SH, Kang JQ, et al. Erythropoietin prevents early and late neuronal demise through modulation of Akt1 and induction of caspase 1, 3, and 8. J Neurosci Res, 2003, 71: 659–669
    4. Villa P, Bigini P, Mennini T, et al. Erythropoietin selectively attenuates cytokine production and inflammation in cerebral ischemia by targeting neuronal apoptosis. J Exp Med, 2003, 198: 971–975.
    5. Genc S, Kuralay F, Genc K, et al. Erythropoietin exerts neuroprotection in 1-methyl-4-phenyl-1, 2,
    3, 6-tetrahydropyridine-treated C57/BL mice via increasing nitric oxide production. Neurosci Lett, 2001, 298: 139–141.
    6. Signore AP, Weng Z, Hastings T, et al. Erythropoietin protects against 6-hydroxydopamine-induced dopaminergic cell death. J Neurochem, 2006, 96: 428-443.
    7. Csete M, Rodriguez L, Wilcox M, et al. Erythropoietin receptor is expressed on adult rat dopaminergic neurons and erythropoietin is neurotrophic in cultured dopaminergic neuroblasts. Neurosci Lett, 2004, 359: 124–126.
    8. Seaton TA, Cooper JM, Schapira AH. Free radical scavengers protect dopaminergic cell lines from apoptosis induced by complex I inhibitors. Brain Res, 1997, 777: 110–118.
    9. Di Monte D, Sandy MS, Ekstrom G, et al. Comparative studies on the mechanisms of paraquat and 1-methyl-4-phenylpyridine (MPP+) cytotoxicity. Biochem Biophys Res Commun, 1986, 137: 303–309.
    10. Cassarino DS, Parks JK, Parker WD Jr, et al. The parkinsonian neurotoxin MPP+ opens the mitochondrial permeability transition pore and releases cytochrome c in isolated mitochondria via an oxidative mechanism. Biochem Biophys Acta, 1999, 1453:49–62.
    11. Masuda S, Nagao M, Takahata K, et al. Functional erythropoietin receptor of the cells with neural characteristics. Comparison with receptor properties of erythroid cells. J Biol Chem, 1993, 268:11208–11216.
    12. Frebel K, Wiese S. Signalling molecules essential for neuronal survival and differentiation. Biochem Soc Trans, 2006, 34:1287-1290.
    13. Hasselblatt M, Ehrenreich H, Siren AL. The brain erythropoietin system and its potential for therapeutic exploitation in brain disease. J Neurosurg Anesthesiol, 2006, 18:132-138.
    14. Zhang F, Signore AP, Zhou Z, et al. Erythropoietin protects CA1 neurons against global cerebral ischemia in rat: potential signaling mechanisms. J Neurosci Res, 2006, 83:1241-1251.
    15. Cross DA, Alessi DR, Cohen P, et al. Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature, 1995, 378: 785–789.
    16. Hajduch E, Alessi DR, Hemmings BA, et al. Constitutive activation of protein kinase B alpha by membrane targeting promotes glucose and system A amino acid transport, protein synthesis, andinactivation of glycogen synthase kinase 3 in L6 muscle cells. Diabetes, 1998, 47:1006–1013.
    17. Eldar-Finkelman. Glycogen syntgase kinase 3: an emerging therapeutic target. Trend Mol Med, 2002, 8:126-132
    18. Shaw M, Cohen P, Alessi DR. Further evidence that the inhibition of glycogen synthase kinase-3beta by IGF-1 is mediated by PDK1/PKBinduced phosphorylation of Ser-9 and not by dephosphorylation of Tyr-216. FEBS Lett, 1997, 416: 307–311.
    19. Pap M, Cooper GM. Role of glycogen synthase kinase-3 in the phosphatidylinositol 3-kinase/Akt cell survival pathway. J Biol Chem, 1998, 273:19929–19932.
    20. Somervaille TC, Linch DC, Khwaja A. Growth factor withdrawal from primary human erythroid progenitors induces apoptosis through a pathway involving glycogen synthase kinase-3 and Bax. Blood, 2001, 98:1374–1381.
    21. Bijur GN, Jope RS. Proapoptotic stimuli induce nuclear accumulation of glycogen synthase kinase-3 beta. J Biol Chem, 2001, 276: 37436–37442.
    22. 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, 919:106–114.
    23. Bo J, Ming BY, Gang LZ, et al. Protection by puerarin against MPP+-induced neurotoxicity in PC12 cells mediated by inhibiting mitochondrial dysfunction and caspase-3-like activation. Neurosci Res, 2005, 53:183–188.
    24. Lee CS, Han ES, Kim YK. Piperine inhibition of 1-methyl-4-phenylpyridinium-induced mitochondrial dysfunction and cell death in PC12 cells. Eur J Pharmacol, 2006, 537: 37–44.
    25. Guan S, Jiang B, Bao YM, et al. Protocatechuic acid suppresses MPP (+)-induced mitochondrial dysfunction and apoptotic cell death in PC12 cells. Food Chem Toxicol, 2006, 44: 1659–1666.
    26. Xia Z, Dickens M, Raingeaud J, et al. Opposing effects of Erk and Jnk-P38 Map kinases on apoptosis. Science, 1995,270:1326–1331.
    27. Ruscher K, Freyer D, Karsch M, et al. Erythropoietin is a paracrine mediator of is ischemic tolerance in the brain: evidence from an in vitro model. J Neurosci, 2002, 22: 10291–10301.
    28. Hanlon PR, Fu P, Wright GL, et al. Mechanisms of erythropoietin-mediated cardioprotection during ischemia-reperfusion injury: role of protein kinase C and phosphatidylinositol 3-kinase signaling. FASEB J, 2005, 19:1323–1325.
    29. Signore AP, Weng Z, Hastings T, et al. Erythropoietin protects against 6-hydroxydopamine-induced dopaminergic cell death. J Neurochem. 2006, 96: 428–443.
    30. Siren AL, Fratelli M, Brines M, et al. Erythropoietin prevents neuronal apoptosis after cerebralischemia and metabolic stress. Proc Natl Acad Sci, 2001, 98:4044–4049.
    31. Lee SM, Nguyen TH, Park MH, et al. EPO receptormediated ERK kinase and NF-kappaB activation in erythropoietin-promoted differentiation of astrocytes. Biochem Biophys Res Commun, 2004, 320: 1087–1095.
    32. Kilic E, Kilic U, Soliz J, et al. Brain-derived erythropoietin protects from focal cerebral ischemia by dual activation of ERK-1/-2 and Akt pathways. FASEB J, 2005, 19: 2026–2028.
    33. Wu DC, Ye W, Che XM, et al. Activation of mitogen-activated protein kinases after permanent cerebral artery occlusion in mouse brain. J Cerebral Blood Flow Metab, 2000, 20: 1320-1330.
    34. Fukunaga K, Miyamoto E. Role of MAP kinase in neurons. Mol Neurobiol, 1998, 16: 79-95.
    35. Xia Z, Dickens M, Raingeaud J, et al. Opposing effects of ERK and JNK-p38 MAP kinases on apoptosis. Science, 1995, 270: 1326-1331.
    36. Grewal SS,York RD,Stork P JS. Extracellular-signal-regulated kinase signalling in neurons. Curr Opin Neurobio, 1999, 9: 544-553.
    37. Harris CA, Johnson EM Jr. BH3-only Bcl-2 family members are coordinately regulated by the JNK pathway and require Bax to induce apoptosis in neurons. J Biol Chem, 2001, 276: 37754-37760.
    38. Gu Z, Jiang Q, Zhang G, et al. Extracellular signal-regulated kinase and c-jun N-terminal protein kinases in ischemic tolerance. Neuroreport, 2001, 12: 3487-3491.
    39. Alessandrini A, Namura S, Moskowitz MA, et al. MEK1 protein kinase inhibition protects against damage resulting from focal cerebral ischemia. Proc Natl Acad Sci USA, 1999, 96: 12866-12869.
    1. Olanow CW, Obeso JA, Stocchi F. Drug insight: Continuous dopaminergic stimulation in the treatment of Parkinson's disease. Nat Clin Pract Neurol. 2006, 2(7):382-92.
    2. Olanow CW, Obeso JA, Stocchi F. Continuous dopamine-receptor treatment of Parkinson's disease: scientific rationale and clinical implications. Lancet Neurol. 2006, 5(8):677-87.
    3. Nyholm D. The rationale for continuous dopaminergic stimulation in advanced Parkinson's disease. Parkinsonism Relat Disord. 2007 [Epub ahead of print]
    4. Stocchi F, Vacca L, Ruggieri S, et al. Intermittent vs continuous levodopa administration in patients with advanced Parkinson disease: a clinical and pharmacokinetic study. Arch Neurol. 2005, 62(6):905-10.
    5. Nilsson D, Hansson LE, Johansson K, et al. Long-term intraduodenal infusion of a water based levodopa-carbidopa dispersion in very advanced Parkinson's disease. Acta Neurol Scand. 1998, 97(3):175-83.
    6. Koller WC, Hutton JT, Tolosa E, et al. Immediate-release and controlled-release carbidopa/levodopa in PD: a 5-year randomized multicenter study. Carbidopa/Levodopa Study Group. Neurology. 1999, 53(5):1012-9.
    7. Stocchi F, Barbato L, Bramante L, et al. Fluctuating parkinsonism: a pilot study of single afternoon dose of levodopa methyl ester. J Neurol. 1996, 243(5):377-80.
    8. Nyholm D. Pharmacokinetic optimisation in the treatment of Parkinson's disease : an update. Clin Pharmacokinet. 2006, 45(2):109-36.
    9. Müller T, Erdmann C, Muhlack S, et al. Inhibition of catechol-O-methyltransferase contributes to more stable levodopa plasma levels. Mov Disord, 2006, 21(3):332-6.
    10. Olanow CW, Stocchi F. COMT inhibitors in Parkinson's disease: can they prevent and/or reverse levodopa-induced motor complications? Neurology. 2004, 62:S72-81.
    11. Schrag A. Entacapone in the treatment of Parkinson's disease. Lancet Neurol. 2005, 4(6):366-70
    12. Smith LA, Jackson MJ, Al-Barghouthy G, et al. Multiple small doses of levodopa plus entacapone produce continuous dopaminergic stimulation and reduce dyskinesia induction in MPTP-treated drug-naive primates. Mov Disord. 2005, 20(3):306-14.
    13. Olanow CW, Kieburtz K, Stern M, et al. Double-blind, placebo-controlled study of entacapone in levodopa-treated patients with stable Parkinson disease. Arch Neurol. 2004, 61(10):1563-8.
    14. Rascol O, Brooks DJ, Korczyn AD, et al. A five-year study of the incidence of dyskinesia in patients with early Parkinson's disease who were treated with ropinirole or levodopa. 056 Study Group. N Engl J Med. 2000, 342(20):1484-91.
    15. Parkinson Study Group. Pramipexole vs levodopa as initial treatment for Parkinson disease: A randomized controlled trial. Parkinson Study Group. JAMA. 2000, 284(15):1931-8.
    16. Katzenschlager R, Hughes A, Evans A, et al. Continuous subcutaneous apomorphine therapy improves dyskinesias in Parkinson's disease: a prospective study using single-dose challenges. Mov Disord. 2005, 20(2):151-7.
    17. LeWitt PA, Lyons KE, Pahwa R. Advanced Parkinson disease treated with rotigotine transdermal system: PREFER Study. Neurology. 2007, 68(16):1262-7.
    18. Woitalla D, Müller T, Benz S, et al. Transdermal lisuride delivery in the treatment of Parkinson's disease. J Neural Transm Suppl. 2004, 68:89-95.
    19. Nutt JG. Continuous dopaminergic stimulation: Is it the answer to the motor complications of Levodopa? Mov Disord. 2007, 22(1):1-9.

© 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