[HTML][HTML] Macrophage-produced VEGFC is induced by efferocytosis to ameliorate cardiac injury and inflammation

KE Glinton, W Ma, C Lantz… - The Journal of …, 2022 - Am Soc Clin Investig
KE Glinton, W Ma, C Lantz, LS Grigoryeva, M DeBerge, X Liu, M Febbraio, M Kahn, G Oliver
The Journal of Clinical Investigation, 2022Am Soc Clin Investig
Clearance of dying cells by efferocytosis is necessary for cardiac repair after myocardial
infarction (MI). Recent reports have suggested a protective role for vascular endothelial
growth factor C (VEGFC) during acute cardiac lymphangiogenesis after MI. Here, we report
that defective efferocytosis by macrophages after experimental MI led to a reduction in
cardiac lymphangiogenesis and Vegfc expression. Cell-intrinsic evidence for efferocytic
induction of Vegfc was revealed after adding apoptotic cells to cultured primary …
Clearance of dying cells by efferocytosis is necessary for cardiac repair after myocardial infarction (MI). Recent reports have suggested a protective role for vascular endothelial growth factor C (VEGFC) during acute cardiac lymphangiogenesis after MI. Here, we report that defective efferocytosis by macrophages after experimental MI led to a reduction in cardiac lymphangiogenesis and Vegfc expression. Cell-intrinsic evidence for efferocytic induction of Vegfc was revealed after adding apoptotic cells to cultured primary macrophages, which subsequently triggered Vegfc transcription and VEGFC secretion. Similarly, cardiac macrophages elevated Vegfc expression levels after MI, and mice deficient for myeloid Vegfc exhibited impaired ventricular contractility, adverse tissue remodeling, and reduced lymphangiogenesis. These results were observed in mouse models of permanent coronary occlusion and clinically relevant ischemia and reperfusion. Interestingly, myeloid Vegfc deficiency also led to increases in acute infarct size, prior to the amplitude of the acute cardiac lymphangiogenesis response. RNA-Seq and cardiac flow cytometry revealed that myeloid Vegfc deficiency was also characterized by a defective inflammatory response, and macrophage-produced VEGFC was directly effective at suppressing proinflammatory macrophage activation. Taken together, our findings indicate that cardiac macrophages promote healing through the promotion of myocardial lymphangiogenesis and the suppression of inflammatory cytokines.
The Journal of Clinical Investigation