Dimethyl fumarate preserves left ventricular infarct integrity following myocardial infarction via modulation of cardiac macrophage and fibroblast oxidative metabolism

AJ Mouton, ER Flynn, SP Moak, NM Aitken… - Journal of molecular and …, 2021 - Elsevier
AJ Mouton, ER Flynn, SP Moak, NM Aitken, ACM Omoto, X Li, AA da Silva, Z Wang…
Journal of molecular and cellular cardiology, 2021Elsevier
Myocardial infarction (MI) is one of the leading causes of mortality and cardiovascular
disease worldwide. MI is characterized by a substantial inflammatory response in the
infarcted left ventricle (LV), followed by transition of quiescent fibroblasts to active
myofibroblasts, which deposit collagen to form the reparative scar. Metabolic shifting
between glycolysis and mitochondrial oxidative phosphorylation (OXPHOS) is an important
mechanism by which these cell types transition towards reparative phenotypes. Thus, we …
Abstract
Myocardial infarction (MI) is one of the leading causes of mortality and cardiovascular disease worldwide. MI is characterized by a substantial inflammatory response in the infarcted left ventricle (LV), followed by transition of quiescent fibroblasts to active myofibroblasts, which deposit collagen to form the reparative scar. Metabolic shifting between glycolysis and mitochondrial oxidative phosphorylation (OXPHOS) is an important mechanism by which these cell types transition towards reparative phenotypes. Thus, we hypothesized that dimethyl fumarate (DMF), a clinically approved anti-inflammatory agent with metabolic actions, would improve post-MI remodeling via modulation of macrophage and fibroblast metabolism. Adult male C57BL/6J mice were treated with DMF (10 mg/kg) for 3–7 days after MI. DMF attenuated LV infarct and non-infarct wall thinning at 3 and 7 days post-MI, and decreased LV dilation and pulmonary congestion at day 7. DMF improved LV infarct collagen deposition, myofibroblast activation, and angiogenesis at day 7. DMF also decreased pro-inflammatory cytokine expression (Tnf) 3 days after MI, and decreased inflammatory markers in macrophages isolated from the infarcted heart (Hif1a, Il1b). In fibroblasts extracted from the infarcted heart at day 3, RNA-Seq analysis demonstrated that DMF promoted an anti-inflammatory/pro-reparative phenotype. By Seahorse analysis, DMF did not affect glycolysis in either macrophages or fibroblasts at day 3, but enhanced macrophage OXPHOS while impairing fibroblast OXPHOS. Our results indicate that DMF differentially affects macrophage and fibroblast metabolism, and promotes anti-inflammatory/pro-reparative actions. In conclusion, targeting cellular metabolism in the infarcted heart may be a promising therapeutic strategy.
Elsevier