[HTML][HTML] MicroRNA-33–dependent regulation of macrophage metabolism directs immune cell polarization in atherosclerosis

M Ouimet, HN Ediriweera, UM Gundra… - The Journal of …, 2015 - Am Soc Clin Investig
M Ouimet, HN Ediriweera, UM Gundra, FJ Sheedy, B Ramkhelawon, SB Hutchison…
The Journal of clinical investigation, 2015Am Soc Clin Investig
Cellular metabolism is increasingly recognized as a controller of immune cell fate and
function. MicroRNA-33 (miR-33) regulates cellular lipid metabolism and represses genes
involved in cholesterol efflux, HDL biogenesis, and fatty acid oxidation. Here, we determined
that miR-33–mediated disruption of the balance of aerobic glycolysis and mitochondrial
oxidative phosphorylation instructs macrophage inflammatory polarization and shapes
innate and adaptive immune responses. Macrophage-specific Mir33 deletion increased …
Cellular metabolism is increasingly recognized as a controller of immune cell fate and function. MicroRNA-33 (miR-33) regulates cellular lipid metabolism and represses genes involved in cholesterol efflux, HDL biogenesis, and fatty acid oxidation. Here, we determined that miR-33–mediated disruption of the balance of aerobic glycolysis and mitochondrial oxidative phosphorylation instructs macrophage inflammatory polarization and shapes innate and adaptive immune responses. Macrophage-specific Mir33 deletion increased oxidative respiration, enhanced spare respiratory capacity, and induced an M2 macrophage polarization–associated gene profile. Furthermore, miR-33–mediated M2 polarization required miR-33 targeting of the energy sensor AMP-activated protein kinase (AMPK), but not cholesterol efflux. Notably, miR-33 inhibition increased macrophage expression of the retinoic acid–producing enzyme aldehyde dehydrogenase family 1, subfamily A2 (ALDH1A2) and retinal dehydrogenase activity both in vitro and in a mouse model. Consistent with the ability of retinoic acid to foster inducible Tregs, miR-33–depleted macrophages had an enhanced capacity to induce forkhead box P3 (FOXP3) expression in naive CD4+ T cells. Finally, treatment of hypercholesterolemic mice with miR-33 inhibitors for 8 weeks resulted in accumulation of inflammation-suppressing M2 macrophages and FOXP3+ Tregs in plaques and reduced atherosclerosis progression. Collectively, these results reveal that miR-33 regulates macrophage inflammation and demonstrate that miR-33 antagonism is atheroprotective, in part, by reducing plaque inflammation by promoting M2 macrophage polarization and Treg induction.
The Journal of Clinical Investigation