Your browser doesn't support javascript.
loading
Microglial LRRK2-mediated NFATc1 attenuates α-synuclein immunotoxicity in association with CX3CR1-induced migration and the lysosome-initiated degradation.
Feng, Linjuan; Lo, Hsuan; Hong, Zhaoxiang; Zheng, Jiahao; Yan, Yuhong; Ye, Zucheng; Chen, Xiaochun; Pan, Xiaodong.
  • Feng L; Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, Fuzhou, China.
  • Lo H; Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China.
  • Hong Z; Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China.
  • Zheng J; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China.
  • Yan Y; Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
  • Ye Z; Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, Fuzhou, China.
  • Chen X; Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China.
  • Pan X; Department of Neurology, The University of HongKong Shenzhen Hospital, Shenzhen, Guangdong, China.
Glia ; 71(9): 2266-2284, 2023 09.
Article en En | MEDLINE | ID: mdl-37300531
ABSTRACT
Synucleinopathies refer to a range of neurodegenerative diseases caused by abnormal α-synuclein (α-Syn) deposition, including Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). Their pathogenesis is strongly linked to microglial dysfunction and neuroinflammation, which involves the leucine-rich-repeat kinase 2 (LRRK2)-regulated nuclear factor of activated T-cells (NFAT). Of the NFAT family, NFATc1 has been found to be increasingly translocated into the nucleus in α-syn stimulation. However, the specific role of NFATc1-mediated intracellular signaling in PD remains elusive in regulating microglial functions. In the current study, we crossbred LRRK2 or NFATc1 conditional knockout mice with Lyz2Cre mice to generate mice with microglia-specific deletion of LRRK2 or NFATc1, and by stereotactic injection of fibrillary α-Syn, we generated PD models in these mice. We found that LRRK2 deficiency enhanced microglial phagocytosis in the mice after α-Syn exposure and that genetic inhibition of NFATc1 markedly diminished phagocytosis and α-Syn elimination. We further demonstrated that LRRK2 negatively regulated NFATc1 in α-Syn-treated microglia, in which microglial LRRK2-deficiency facilitated NFATc1 nuclear translocation, CX3CR1 upregulation, and microglia migration. Additionally, NFATc1 translocation upregulated the expression of Rab7 and promoted the formation of late lysosomes, resulting in α-Syn degradation. In contrast, the microglial NFATc1 deficiency impaired CX3CR1 upregulation and the formation of Rab7-mediated late lysosomes. These findings highlight the critical role of NFATc1 in modulating microglial migration and phagocytosis, in which the LRRK2-NFATc1 signaling pathway regulates the expression of microglial CX3CR1 and endocytic degradative Rab7 to attenuate α-synuclein immunotoxicity.
Asunto(s)
Palabras clave

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Enfermedad de Parkinson / Alfa-Sinucleína Tipo de estudio: Prognostic_studies / Risk_factors_studies Límite: Animals Idioma: En Año: 2023 Tipo del documento: Article

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Enfermedad de Parkinson / Alfa-Sinucleína Tipo de estudio: Prognostic_studies / Risk_factors_studies Límite: Animals Idioma: En Año: 2023 Tipo del documento: Article