Unknown

Dataset Information

0

Disrupting LXR? phosphorylation promotes FoxM1 expression and modulates atherosclerosis by inducing macrophage proliferation.


ABSTRACT: Macrophages are key immune cells for the initiation and development of atherosclerotic lesions. However, the macrophage regulatory nodes that determine how lesions progress in response to dietary challenges are not fully understood. Liver X receptors (LXRs) are sterol-regulated transcription factors that play a central role in atherosclerosis by integrating cholesterol homeostasis and immunity. LXR pharmacological activation elicits a robust antiatherosclerotic transcriptional program in macrophages that can be affected by LXR? S196 phosphorylation in vitro. To investigate the impact of these transcriptional changes in atherosclerosis development, we have generated mice carrying a Ser-to-Ala mutation in myeloid cells in the LDL receptor (LDLR)-deficient atherosclerotic background (M-S196ALdlr-KO). M-S196ALdlr-KO mice fed a high-fat diet exhibit increased atherosclerotic plaque burden and lesions with smaller necrotic cores and thinner fibrous caps. These diet-induced phenotypic changes are consistent with a reprogramed macrophage transcriptome promoted by LXR?-S196A during atherosclerosis development. Remarkably, expression of several proliferation-promoting factors, including the protooncogene FoxM1 and its targets, is induced by LXR?-S196A. This is consistent with increased proliferation of plaque-resident cells in M-S196ALdlr-KO mice. Moreover, disrupted LXR? phosphorylation increases expression of phagocytic molecules, resulting in increased apoptotic cell removal by macrophages, explaining the reduced necrotic cores. Finally, the macrophage transcriptome promoted by LXR?-S196A under dietary perturbation is markedly distinct from that revealed by LXR ligand activation, highlighting the singularity of this posttranslational modification. Overall, our findings demonstrate that LXR? phosphorylation at S196 is an important determinant of atherosclerotic plaque development through selective changes in gene transcription that affect multiple pathways.

SUBMITTER: Gage MC 

PROVIDER: S-EPMC6048484 | biostudies-literature | 2018 Jul

REPOSITORIES: biostudies-literature

altmetric image

Publications

Disrupting LXRα phosphorylation promotes FoxM1 expression and modulates atherosclerosis by inducing macrophage proliferation.

Gage M C MC   Bécares N N   Louie R R   Waddington K E KE   Zhang Y Y   Tittanegro T H TH   Rodríguez-Lorenzo S S   Jathanna A A   Pourcet B B   Pello O M OM   De la Rosa J V JV   Castrillo A A   Pineda-Torra I I  

Proceedings of the National Academy of Sciences of the United States of America 20180627 28


Macrophages are key immune cells for the initiation and development of atherosclerotic lesions. However, the macrophage regulatory nodes that determine how lesions progress in response to dietary challenges are not fully understood. Liver X receptors (LXRs) are sterol-regulated transcription factors that play a central role in atherosclerosis by integrating cholesterol homeostasis and immunity. LXR pharmacological activation elicits a robust antiatherosclerotic transcriptional program in macroph  ...[more]

Similar Datasets

| S-SCDT-10_15252-EMMM_202217198 | biostudies-other
2023-02-27 | PXD039083 | Pride
2016-05-06 | GSE67014 | GEO
2022-12-24 | GSE221647 | GEO
2016-05-06 | GSE67011 | GEO
2016-05-06 | GSE67013 | GEO
2024-03-25 | PXD050932 | Pride
| S-EPMC3769444 | biostudies-literature
| S-EPMC4619168 | biostudies-literature
| S-EPMC4878149 | biostudies-literature