Atherosclerosis-Driven Treg Plasticity Results in Formation of a Dysfunctional Subset of Plastic IFNγ+ Th1/Tregs

MJ Butcher, AR Filipowicz, TC Waseem… - Circulation …, 2016 - Am Heart Assoc
MJ Butcher, AR Filipowicz, TC Waseem, CM McGary, KJ Crow, N Magilnick, M Boldin…
Circulation research, 2016Am Heart Assoc
Rationale: Forkhead box P3+ T regulatory cells (Tregs) are key players in maintaining
immune homeostasis. Evidence suggests that Tregs respond to environmental cues to
permit or suppress inflammation. In atherosclerosis, Th1-driven inflammation affects Treg
homeostasis, but the mechanisms governing this phenomenon are unclear. Objective: Here,
we address whether atherosclerosis impacts Treg plasticity and functionality in Apoe−/−
mice, and what effect Treg plasticity might have on the pathology of atherosclerosis …
Rationale:
Forkhead box P3+ T regulatory cells (Tregs) are key players in maintaining immune homeostasis. Evidence suggests that Tregs respond to environmental cues to permit or suppress inflammation. In atherosclerosis, Th1-driven inflammation affects Treg homeostasis, but the mechanisms governing this phenomenon are unclear.
Objective:
Here, we address whether atherosclerosis impacts Treg plasticity and functionality in Apoe/− mice, and what effect Treg plasticity might have on the pathology of atherosclerosis.
Methods and Results:
We demonstrate that atherosclerosis promotes Treg plasticity, resulting in the reduction of CXCR3+ Tregs and the accumulation of an intermediate Th1-like interferon (IFN)-γ+CCR5+ Treg subset (Th1/Tregs) within the aorta. Importantly, Th1/Tregs arise in atherosclerosis from bona fide Tregs, rather than from T-effector cells. We show that Th1/Tregs recovered from atherosclerotic mice are dysfunctional in suppression assays. Using an adoptive transfer system and plasticity-prone Mir146a−/− Tregs, we demonstrate that elevated IFNγ+ Mir146a−/− Th1/Tregs are unable to adequately reduce atherosclerosis, arterial Th1, or macrophage content within Apoe−/− mice, in comparison to Mir146a+/+ Tregs. Finally, via single-cell RNA-sequencing and real-time -polymerase chain reaction, we show that Th1/Tregs possess a unique transcriptional phenotype characterized by coexpression of Treg and Th1 lineage genes and a downregulation of Treg-related genes, including Ikzf2, Ikzf4, Tigit, Lilrb4, and Il10. In addition, an ingenuity pathway analysis further implicates IFNγ, IFNα, interleukin-2, interleukin-7, CTLA-4 (cytotoxic T-lymphocyte–associated protein 4), T-cell receptor, and Csnk2b-related pathways in regulating Treg plasticity.
Conclusions:
Atherosclerosis drives Treg plasticity, resulting in the accumulation of dysfunctional IFNγ+ Th1/Tregs that may permit further arterial inflammation and atherogenesis.
Am Heart Assoc