期刊文献+

Regulation of adenosine triphosphate-sensitive potassium channels suppresses the toxic effects of amyloid-beta peptide(25-35)

Regulation of adenosine triphosphate-sensitive potassium channels suppresses the toxic effects of amyloid-beta peptide(25-35)
下载PDF
导出
摘要 In this study, we treated PC12 cells with 0-20 μM amyloid-β peptide (25-35) for 24 hours to induce cytotoxicity, and found that 5-20 μM amyloid-β peptide (25-35) decreased PC12 cell viability, but adenosine triphosphate-sensitive potassium channel activator diazoxide suppressed the decrease in PC12 cell viability induced by amyloid-β peptide (25-35). Diazoxide protected PC12 cells against amyloid-β peptide (25-35)-induced increases in mitochondrial membrane potential and intracellular reactive oxygen species levels. These protective effects were reversed by the selective mitochondrial adenosine triphosphate-sensitive potassium channel blocker 5-hydroxydecanoate. An inducible nitric oxide synthase inhibitor, Nw-nitro-L-arginine, also protected PC12 cells from amyloid-β peptide (25-35)-induced increases in both mitochondrial membrane potential and intracellular reactive oxygen species levels. However, the H202-degrading enzyme catalase could not reverse the amyloid-β peptide (25-35)-induced increase in intracellular reactive oxygen species. A 24-hour exposure to amyloid-13 peptide (25-35) did not result in apoptosis or necrosis, suggesting that the increases in both mitochondrial membrane potential and reactive oxygen species levels preceded cell death. The data suggest that amyloid-β peptide (25-35) cytotoxicity is associated with adenosine triphosphate-sensitive potassium channels and nitric oxide. Regulation of adenosine triphosphate-sensitive potassium channels suppresses PC12 cell cytotoxicity induced by amyloid-β peptide (25-35). In this study, we treated PC12 cells with 0-20 μM amyloid-β peptide (25-35) for 24 hours to induce cytotoxicity, and found that 5-20 μM amyloid-β peptide (25-35) decreased PC12 cell viability, but adenosine triphosphate-sensitive potassium channel activator diazoxide suppressed the decrease in PC12 cell viability induced by amyloid-β peptide (25-35). Diazoxide protected PC12 cells against amyloid-β peptide (25-35)-induced increases in mitochondrial membrane potential and intracellular reactive oxygen species levels. These protective effects were reversed by the selective mitochondrial adenosine triphosphate-sensitive potassium channel blocker 5-hydroxydecanoate. An inducible nitric oxide synthase inhibitor, Nw-nitro-L-arginine, also protected PC12 cells from amyloid-β peptide (25-35)-induced increases in both mitochondrial membrane potential and intracellular reactive oxygen species levels. However, the H202-degrading enzyme catalase could not reverse the amyloid-β peptide (25-35)-induced increase in intracellular reactive oxygen species. A 24-hour exposure to amyloid-13 peptide (25-35) did not result in apoptosis or necrosis, suggesting that the increases in both mitochondrial membrane potential and reactive oxygen species levels preceded cell death. The data suggest that amyloid-β peptide (25-35) cytotoxicity is associated with adenosine triphosphate-sensitive potassium channels and nitric oxide. Regulation of adenosine triphosphate-sensitive potassium channels suppresses PC12 cell cytotoxicity induced by amyloid-β peptide (25-35).
出处 《Neural Regeneration Research》 SCIE CAS CSCD 2013年第1期56-63,共8页 中国神经再生研究(英文版)
基金 supported by the Project Sponsored by Yantai Science and Technology Bureau,China,No.2010232
关键词 neural regeneration neurodegenerative diseases amyloid-β peptide (25-35) PC12 cell adenosinetriphosphate-sensitive potassium channel inducible nitric oxide synthase mitochondrial membranepotential reactive oxygen species grant-supported paper photographs-containing paper NEUROREGENERATION neural regeneration neurodegenerative diseases amyloid-β peptide (25-35) PC12 cell adenosinetriphosphate-sensitive potassium channel inducible nitric oxide synthase mitochondrial membranepotential reactive oxygen species grant-supported paper photographs-containing paper neuroregeneration
  • 相关文献

参考文献31

  • 1Lehéricy S,Hirsch EC,Cervera-Piérot P. Heterogeneity and selectivity of the degeneration of cholinergic neurons in the basal forebrain of patients with Alzheimer's disease[J].Journal of Comparative Neurology,1993,(01):15-31.
  • 2Barnham KJ,Cappai R,Beyreuther K. Delineating common molecular mechanisms in Alzheimer's and prion diseases[J].Trends,.
  • 3Abad MA,Enguita M,DeGregorio-Rocasolano N. Neuronal pentraxin 1 contributes to the neuronal damage evoked by amyloid-beta and is overexpressed in dystrophic neurites in Alzheimer's brain[J].The Journal of Neuroscience,2006,(49):12735-12747.
  • 4Tickler AK,Wade JD,Separovic F. The role of Abeta peptides in Alzheimer's disease[J].Protein and Peptide Letters,2005,(06):513-519.doi:10.2174/0929866054395905.
  • 5Kaminsky YG,Marlatt MW,Smith MA. Subcellular and metabolic examination of amyloid-beta peptides in Alzheimer disease pathogenesis:evidence for Abeta(25-35)[J].Experimental Neurology,2010,(01):26-37.
  • 6Gao X,Tang XC. Huperzine A attenuates mitochondrial dysfunction in beta-amyloid-treated PC12 cells by reducing oxygen free radicals accumulation and improving mitochondrial energy metabolism[J].Journal of Neuroscience Research,2006,(06):1048-1057.
  • 7Silva DF,Esteves AR,Oliveira CR. Mitochondria:the common upstream driver of amyloid-β and tau pathology in Alzheimer's disease[J].Current Alzheimer Research,2011,(05):563-572.
  • 8Maruszak A,(Z)ekanowski C. Mitochondrial dysfunction and Alzheimer's disease[J].Progress in Neuro-Psychopharmacology & Biological Psychiatry,2011,(02):320-330.
  • 9Zawar C,Plant TD,Schirra C. Cell-type specific expression of ATP-sensitive potassium channels in the rat hippocampus[J].The Journal of Physiology,1999,(Pt 2):327-341.
  • 10Ma G,Fu Q,Zhang Y. Effects of Abeta1-42 on the subunits of KATP expression in cultured primary rat basal forebrain neurons[J].Neurochemical Research,2008,(07):1419-1424.doi:10.1007/s11064-008-9603-2.

相关作者

内容加载中请稍等...

相关机构

内容加载中请稍等...

相关主题

内容加载中请稍等...

浏览历史

内容加载中请稍等...
;
使用帮助 返回顶部