1. Academic Validation
  2. p38-TFEB pathways promote microglia activation through inhibiting CMA-mediated NLRP3 degradation in Parkinson's disease

p38-TFEB pathways promote microglia activation through inhibiting CMA-mediated NLRP3 degradation in Parkinson's disease

  • J Neuroinflammation. 2021 Dec 20;18(1):295. doi: 10.1186/s12974-021-02349-y.
Jialong Chen  # 1 2 Kanmin Mao  # 1 Honglin Yu  # 1 Yue Wen 1 Hua She 3 He Zhang 2 Linhua Liu 2 Mingque Li 2 Wenjun Li 4 Fei Zou 5
Affiliations

Affiliations

  • 1 Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, No. 1838, North Guangzhou Road, Guangzhou, 510515, Guangdong Province, China.
  • 2 School of Public Health, Guangdong Medical University, Dongguan,, Guangdong Province, China.
  • 3 Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, USA.
  • 4 Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, No. 1838, North Guangzhou Road, Guangzhou, 510515, Guangdong Province, China. [email protected].
  • 5 Department of Occupational Health and Occupational Medicine, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, No. 1838, North Guangzhou Road, Guangzhou, 510515, Guangdong Province, China. [email protected].
  • # Contributed equally.
Abstract

Background: Parkinson's disease (PD) is characterized by degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc), accompanied by accumulation of α-synuclein, chronic neuroinflammation and Autophagy dysfunction. Previous studies suggested that misfolded α-synuclein induces the inflammatory response and Autophagy dysfunction in microglial cells. The NLRP3 inflammasome signaling pathway plays a crucial role in the neuroinflammatory process in the central nervous system. However, the relationship between Autophagy deficiency and NLRP3 activation induced by α-synuclein accumulation is not well understood.

Methods: Through immunoblotting, immunocytochemistry, immunofluorescence, flow cytometry, ELISA and behavioral tests, we investigated the role of p38-TFEB-NLRP3 signaling pathways on neuroinflammation in the α-synuclein A53T PD models.

Results: Our results showed that increased protein levels of NLRP3, ASC, and Caspase-1 in the α-synuclein A53T PD models. P38 is activated by overexpression of α-synuclein A53T mutant, which inhibited the master transcriptional activator of Autophagy TFEB. And we found that NLRP3 was degraded by chaperone-mediated Autophagy (CMA) in microglial cells. Furthermore, p38-TFEB pathways inhibited CMA-mediated NLRP3 degradation in Parkinson's disease. Inhibition of p38 had a protective effect on Parkinson's disease model via suppressing the activation of NLRP3 inflammasome pathway. Moreover, both p38 inhibitor SB203580 and NLRP3 Inhibitor MCC950 not only prevented neurodegeneration in vivo, but also alleviated movement impairment in α-synuclein A53T-tg mice model of Parkinson's disease.

Conclusion: Our research reveals p38-TFEB pathways promote microglia activation through inhibiting CMA-mediated NLRP3 degradation in Parkinson's disease, which could be a potential therapeutic strategy for PD. p38-TFEB pathways promote microglia activation through inhibiting CMA-mediated NLRP3 degradation in Parkinson's disease. In this model, p38 activates NLRP3 inflammasome via inhibiting TFEB in microglia. TFEB signaling negatively regulates NLRP3 inflammasome through increasing LAMP2A expression, which binds to NLRP3 and promotes its degradation via chaperone-mediated Autophagy (CMA). NLRP3-mediated microglial activation promotes the death of dopaminergic neurons.

Keywords

Chaperone-mediated autophagy; NLRP3; Parkinson’s disease; TFEB; p38.

Figures
Products