Dysfunctional and Pro-Inflammatory Regulatory T-Lymphocytes are Essential for Adverse Cardiac Remodeling in Ischemic Cardiomyopathy

Sumanth S., Bansal, Shyam S.; Ismahil, Mohamed Ameen; Goel, Mehak; Zhou, Guihua; Rokosh, Gregg; Hamid, Tariq; Prabhu

Circulation |

Background: Heart failure (HF) is a state of inappropriately sustained inflammation, suggesting the loss of normal immunosuppressive mechanisms. Regulatory T-lymphocytes (Tregs) are considered key suppressors of immune responses; however, their role in HF is unknown. We hypothesized that Tregs are dysfunctional in ischemic cardiomyopathy and HF, and promote immune activation and left ventricular (LV) remodeling. Methods: Adult male wild-type (WT) C57BL/6 mice, Foxp3-diptheria toxin receptor(DTR) transgenic mice, and tumor necrosis factor(TNF)α receptor-1(TNFR1)-/- mice underwent non-reperfused myocardial infarction (MI) to induce HF, or sham operation. LV remodeling was assessed by echocardiography, and histological and molecular phenotyping. Alterations in Treg profile and function were examined by flow cytometry, immunostaining, and in vitro cell assays. Results: As compared with WT sham mice, CD4+Foxp3+ Tregs in WT HF mice robustly expanded in the heart, circulation, spleen, and lymph nodes in a phasic manner after MI, beyond the early phase of wound healing, and exhibited pro-inflammatory Th1-type features with interferon-γ, TNFα, and TNFR1 expression, loss of immunomodulatory capacity, heightened proliferation, and potentiated anti-angiogenic and pro-fibrotic properties. Selective Treg ablation in Foxp3-DTR mice with ischemic cardiomyopathy reversed LV remodeling and dysfunction, alleviating hypertrophy and fibrosis, while suppressing circulating CD4+ T-cells and systemic inflammation, and enhancing tissue neovascularization. Importantly, Tregs reconstituted after ablation exhibited restoration of immunosuppressive capacity and normalized TNFR1 expression. Treg dysfunction was also tightly coupled to Treg-endothelial cell contact- and TNFR1-dependent inhibition of angiogenesis, and the mobilization and tissue infiltration of CD34+Flk1+ circulating angiogenic cells in a CCL5/CCR5-dependent manner. Anti-CD25-mediated Treg depletion in WT mice imparted similar benefits on LV remodeling, CACs, and tissue neovascularization. Conclusions: Pro-inflammatory and anti-angiogenic Tregs play an essential pathogenetic role in chronic ischemic HF to promote immune activation and pathological LV remodeling. The restoration of normal Treg function may be a viable approach to therapeutic immunomodulation in this disease.