Back to Search Start Over

Adenosine A2A Receptor in Bone Marrow-Derived Cells Mediated Macrophages M2 Polarization via PPARγ-P65 Pathway in Chronic Hypoperfusion Situation.

Authors :
Mou, Ke-Jie
Shen, Kai-Feng
Li, Yan-Ling
Wu, Zhi-Feng
Duan, Wei
Source :
Frontiers in Aging Neuroscience; 1/3/2022, Vol. 13, p1-14, 14p
Publication Year :
2022

Abstract

Background: The role of adenosine A<subscript>2A</subscript> receptor (A<subscript>2A</subscript>R) in the ischemic white matter damage induced by chronic cerebral hypoperfusion remains obscure. Here we investigated the role of A<subscript>2A</subscript>R in the process of macrophage polarizations in the white matter damage induced by chronic cerebral hypoperfusion and explored the involved signaling pathways. Methods: We combined mouse model and macrophage cell line for our study. White matter lesions were induced in A<subscript>2A</subscript>R knockout mice, wild-type mice, and chimeric mice generated by bone marrow cells transplantation through bilateral common carotid artery stenosis. Microglial/macrophage polarization in the corpus callosum was detected by immunofluorescence. For the cell line experiments, RAW264.7 macrophages were treated with the A<subscript>2A</subscript>R agonist CHS21680 or A<subscript>2A</subscript>R antagonist SCH58261 for 30 min and cultured under low-glucose and hypoxic conditions. Macrophage polarization was examined by immunofluorescence. The expression of peroxisome proliferator activated receptor gamma (PPARγ) and transcription factor P65 was examined by western blotting and real-time polymerase chain reaction (RT-PCR). Inflammatory cytokine factors were assessed by enzyme-linked immunosorbent assay (ELISA) and RT-PCR. Results: Both global A<subscript>2A</subscript>R knockout and inactivation of A<subscript>2A</subscript>R in bone marrow-derived cells enhanced M1 marker expression in chronic ischemic white matter lesions. Under low-glucose and hypoxic conditions, CGS21680 treatment promoted macrophage M2 polarization, increased the expression of PPARγ, P65, and interleukin-10 (IL-10) and suppressed the expression of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β). The CGS21680-induced upregulation of P65 and IL-10 was abolished in macrophages upon PPARγ knockdown. The downregulation of TNF-α and IL-1β by CGS21680 was less affected by PPARγ knockdown. Conclusions: In the cerebral hypoperfusion induced white matter damage, A<subscript>2A</subscript>R signaling in bone marrow-derived cells induces macrophage M2 polarization and increases the expression of the anti-inflammatory factor IL-10 via the PPARγ-P65 pathway, both of which might explain its neuroprotective effect. [ABSTRACT FROM AUTHOR]

Details

Language :
English
ISSN :
16634365
Volume :
13
Database :
Complementary Index
Journal :
Frontiers in Aging Neuroscience
Publication Type :
Academic Journal
Accession number :
154478549
Full Text :
https://doi.org/10.3389/fnagi.2021.792733