Impaired Autophagy in CD11b+ Dendritic Cells Expands CD4+ Regulatory T Cells and Limits Atherosclerosis in Mice

M Clement, J Raffort, F Lareyre, D Tsiantoulas… - Circulation …, 2019 - Am Heart Assoc
M Clement, J Raffort, F Lareyre, D Tsiantoulas, S Newland, Y Lu, L Masters, J Harrison…
Circulation Research, 2019Am Heart Assoc
Rationale: Atherosclerosis is a chronic inflammatory disease. Recent studies have shown
that dysfunctional autophagy in endothelial cells, smooth muscle cells, and macrophages,
plays a detrimental role during atherogenesis, leading to the suggestion that autophagy-
stimulating approaches may provide benefit. Objective: Dendritic cells (DCs) are at the
crossroad of innate and adaptive immune responses and profoundly modulate the
development of atherosclerosis. Intriguingly, the role of autophagy in DC function during …
Rationale
Atherosclerosis is a chronic inflammatory disease. Recent studies have shown that dysfunctional autophagy in endothelial cells, smooth muscle cells, and macrophages, plays a detrimental role during atherogenesis, leading to the suggestion that autophagy-stimulating approaches may provide benefit.
Objective
Dendritic cells (DCs) are at the crossroad of innate and adaptive immune responses and profoundly modulate the development of atherosclerosis. Intriguingly, the role of autophagy in DC function during atherosclerosis and how the autophagy process would impact disease development has not been addressed.
Methods and Results
Here, we show that the autophagic flux in atherosclerosis-susceptible Ldlr−/− (low-density lipoprotein receptor–deficient) mice is substantially higher in splenic and aortic DCs compared with macrophages and is further activated under hypercholesterolemic conditions. RNA sequencing and functional studies on selective cell populations reveal that disruption of autophagy through deletion of Atg16l1 differentially affects the biology and functions of DC subsets in Ldlr−/− mice under high-fat diet. Atg16l1 deficient CD11b+ DCs develop a TGF (transforming growth factor)-β–dependent tolerogenic phenotype and promote the expansion of regulatory T cells, whereas no such effects are seen with Atg16l1 deficient CD8α+ DCs. Atg16l1 deletion in DCs (all CD11c-expressing cells) expands aortic regulatory T cells in vivo, limits the accumulation of T helper cells type 1, and reduces the development of atherosclerosis in Ldlr−/− mice. In contrast, no such effects are seen when Atg16l1 is deleted selectively in conventional CD8α+ DCs and CD103+ DCs. Total T-cell or selective regulatory T-cell depletion abrogates the atheroprotective effect of Atg16l1 deficient DCs.
Conclusions
In contrast to its proatherogenic role in macrophages, autophagy disruption in DCs induces a counter-regulatory response that maintains immune homeostasis in Ldlr−/− mice under high-fat diet and limits atherogenesis. Selective modulation of autophagy in DCs could constitute an interesting therapeutic target in atherosclerosis.
Am Heart Assoc
以上显示的是最相近的搜索结果。 查看全部搜索结果