Your browser doesn't support javascript.
loading
FoxG1 as a Potential Therapeutic Target for Alzheimer's Disease: Modulating NLRP3 Inflammasome via AMPK/mTOR Autophagy Pathway.
Yun, Qi; Ma, Si-Fei; Zhang, Wei-Ning; Gu, Meng; Wang, Jia.
Afiliação
  • Yun Q; Changzhou Children's Hospital Affiliated to Nantong University, 958 Zhongwu Avenue, Changzhou, 213000, Jiangsu Province, China.
  • Ma SF; Changzhou Blood Center, 118 Canal Road, Changzhou, 213000, Jiangsu Province, China.
  • Zhang WN; Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 213000, Jiangsu Province, China.
  • Gu M; Changzhou Children's Hospital Affiliated to Nantong University, 958 Zhongwu Avenue, Changzhou, 213000, Jiangsu Province, China. 1041152404@qq.com.
  • Wang J; Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 213000, Jiangsu Province, China. wangjia@ujs.edu.cn.
Cell Mol Neurobiol ; 44(1): 35, 2024 Apr 17.
Article em En | MEDLINE | ID: mdl-38630150
ABSTRACT
An increasing body of research suggests that promoting microglial autophagy hinders the neuroinflammation initiated though the NLRP3 inflammasome activation in Alzheimer's disease (AD). The function of FoxG1, a crucial transcription factor involved in cell survival by regulating mitochondrial function, remains unknown during the AD process and neuroinflammation occurs. In the present study, we firstly found that Aß peptides induced AD-like neuroinflammation upregulation and downregulated the level of autophagy. Following low-dose Aß25-35 stimulation, FoxG1 expression and autophagy exhibited a gradual increase. Nevertheless, with high-concentration Aß25-35 treatment, progressive decrease in FoxG1 expression and autophagy levels as the concentration of Aß25-35 escalated. In addition, FoxG1 has a positive effect on cell viability and autophagy in the nervous system. In parallel with the Aß25-35 stimulation, we employed siRNA to decrease the expression of FoxG1 in N2A cells. A substantial reduction in autophagy level (Beclin1, LC3II, SQSTM1/P62) and a notable growth in inflammatory response (NLRP3, TNF-α, and IL-6) were observed. In addition, we found FoxG1 overexpression owned the effect on the activation of AMPK/mTOR autophagy pathway and siRNA-FoxG1 successfully abolished this effect. Lastly, FoxG1 suppressed the NLRP3 inflammasome and enhanced the cognitive function in AD-like mouse model induced by Aß25-35. Confirmed by cellular and animal experiments, FoxG1 suppressed NLRP3-mediated neuroinflammation, which was strongly linked to autophagy regulated by AMPK/mTOR. Taken together, FoxG1 may be a critical node in the pathologic progression of AD and has the potential to serve as therapeutic target.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Fatores de Transcrição Forkhead / Inflamassomos / Doença de Alzheimer Idioma: En Ano de publicação: 2024 Tipo de documento: Article País de afiliação: China

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Fatores de Transcrição Forkhead / Inflamassomos / Doença de Alzheimer Idioma: En Ano de publicação: 2024 Tipo de documento: Article País de afiliação: China