期刊文献+

Amyloid 13 Protein Aggravates Neuronal Senescence an, d Cognitive Deficits in 5XFAD Mouse Model of Alzheimer s Disease 被引量:7

Amyloid 13 Protein Aggravates Neuronal Senescence an, d Cognitive Deficits in 5XFAD Mouse Model of Alzheimer s Disease
原文传递
导出
摘要 Background: Amyloid β (Aβ) has been established as a key factor for the pathological changes in the brains of patients with Alzheimer's disease (AD), and cellular senescence is closely associated with aging and cognitive impairment. However, it remains blurred whether, in theAD brains, Aβ accelerates the neuronal senescence and whether this senescence, in turn, impairs the cognitive function. This study aimed to explore the expression of senescence-associated genes in the hippocampal tissue from young to aged 5XFAD mice and their age-matched wild type (WT) mice to determine whether senescent neurons are present in the transgenic AD mouse model. Methods: The 5XFAD mice and age-matched wild type mice, both raised from 1 to 18 months, were enrolled in the study. The senescence-associated genes in the hippocampus were analyzed and differentially expressed genes (DEGs) were screened by quantitative real-time polymerase chain reaction. Cognitive perfom3ance of the mice was evaluated by Y-maze and Morris water maze tests. Oligomeric Aβ(oAβ) (1-42) was applied to culture primary neurons to simulate the in vivo manifestation. Aging-related proteins were detected by Western blotting analysis and immunofluorescence. Results: In 5XFAD mice, of all the DEGs, the senescence-associated marker p16 was most significantly increased, even at the early age. It was mainly localized in neurons, with a marginal expression in astrocytes (labeled as glutamine synthetase), nil expression in activated microglia (labeled as lba1), and negatively correlated with the spatial cognitive impairments of 5XFAD mice. oAβ (1-42) induced the production of senescence-related protein p1 6, but not p53 in vitro, which was in line with the in vivo manifestation. Conclusions: oAβ-accelerated neuronal senescence may be associated with the cognitive impairment in 5XFAD mice. Senescence-associated marker p16 can serve as an indicator to estimate the cognitive prognosis for A D population. Background: Amyloid β (Aβ) has been established as a key factor for the pathological changes in the brains of patients with Alzheimer's disease (AD), and cellular senescence is closely associated with aging and cognitive impairment. However, it remains blurred whether, in theAD brains, Aβ accelerates the neuronal senescence and whether this senescence, in turn, impairs the cognitive function. This study aimed to explore the expression of senescence-associated genes in the hippocampal tissue from young to aged 5XFAD mice and their age-matched wild type (WT) mice to determine whether senescent neurons are present in the transgenic AD mouse model. Methods: The 5XFAD mice and age-matched wild type mice, both raised from 1 to 18 months, were enrolled in the study. The senescence-associated genes in the hippocampus were analyzed and differentially expressed genes (DEGs) were screened by quantitative real-time polymerase chain reaction. Cognitive perfom3ance of the mice was evaluated by Y-maze and Morris water maze tests. Oligomeric Aβ(oAβ) (1-42) was applied to culture primary neurons to simulate the in vivo manifestation. Aging-related proteins were detected by Western blotting analysis and immunofluorescence. Results: In 5XFAD mice, of all the DEGs, the senescence-associated marker p16 was most significantly increased, even at the early age. It was mainly localized in neurons, with a marginal expression in astrocytes (labeled as glutamine synthetase), nil expression in activated microglia (labeled as lba1), and negatively correlated with the spatial cognitive impairments of 5XFAD mice. oAβ (1-42) induced the production of senescence-related protein p1 6, but not p53 in vitro, which was in line with the in vivo manifestation. Conclusions: oAβ-accelerated neuronal senescence may be associated with the cognitive impairment in 5XFAD mice. Senescence-associated marker p16 can serve as an indicator to estimate the cognitive prognosis for A D population.
出处 《Chinese Medical Journal》 SCIE CAS CSCD 2016年第15期1835-1844,共10页 中华医学杂志(英文版)
基金 We would like to thank teacher Hong-Zhi Huang from College of Foreign Languages of Fujian Medical University for the kindly help of text modifications and retouching of this paper. This study was supported by grants from National Natural Science Fotmdation of China (No. 81200991 and No. 81571257), Fujian Provincial Natural Science Foundation (No. 2015J01398), Fujian Provincial New Century Excellent Talents Support Program, China (JA13131), and Young and Middle-aged Talent Training Key Project in Health System of Fujian Province (2014-ZQN-ZD- 11).
关键词 Alzheimer's Disease Amyloid β COGNITION P 16 SENESCENCE Alzheimer's Disease Amyloid β Cognition P 16 Senescence
  • 相关文献

参考文献53

  • 1Querfurth HW, LaFerla FM. Alzheimer's disease. N Engl J Med 2010;362:329-44. doi: 10.1056/NEJMra0909142.
  • 2Campisi J. Aging, cellular senescence, and cancer. Annu Rev Physiol 2013;75:685-705. doi: 10.1146/annurev-physiol-030212-183653.
  • 3Deak F, Freeman WM, Ungvari Z, Csiszar A, Sonntag WE. Recent developments in understanding brain aging: Implications for Alzheimer's disease and vascular cognitive impairment. J Gerontol A Biol Sci Med Sci 2016;71:13-20. doi: 10.1093/gerona/glv206.
  • 4L6pez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell 2013;153:1194-217. doi: 10.1016/j. cell.2013.05.039.
  • 5Tacutu R, Budovsky A, Yanai H, Fraifeld VE. Molecular links between cellular senescence, longevity and age-related diseases - A systems biology perspective. Aging (Albany NY) 2011;3:1178-91. doi: 10.18632/aging. 100413.
  • 6Campisi J, Andersen JK, Kapahi P, Melov S. Cellular senescence: A link between cancer and age-related degenerative disease? Semin Cancer Biol 2011 ;21:354-9. doi: 10.1016/j.semeancer.2011.09.001.
  • 7Campisi J. Cellular senescence: Putting the paradoxes in perspective. Curr Opin Genet Dev 2011;21:107-12. doi: 10.1016/j. gde.2010.10.005.
  • 8Kuilman T, Michaloglou C, Mooi WJ, Peeper DS. The essence of senescence. Genes Dev 2010;24:2463-79. doi: 10.1101/gad. 1971610.
  • 9Zhan Y, Song C, Karlsson R, Tillander A, Reynolds CA, Pedersen NL, et al. Telomere length shortening and Alzheimer disease - A Mendelian Randomization Study. JAMA Neurol 2015;72:1202-3. doi: 10.1001/jamaneurol.2015.1513.
  • 10Mombach JC, Bugs CA, Chaouiya C. Modelling the onset of senescence at the G1/S cell cycle checkpoint. BMC Genomics 2014;15 Suppl 7:$7. doi: 10.1186/1471-2164-15-$7-$7.

同被引文献26

引证文献7

二级引证文献31

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