Lymphatic and immune cell cross-talk regulates cardiac recovery after experimental myocardial infarction

M Houssari, A Dumesnil, V Tardif, R Kivelä… - … , and vascular biology, 2020 - Am Heart Assoc
M Houssari, A Dumesnil, V Tardif, R Kivelä, N Pizzinat, I Boukhalfa, D Godefroy…
Arteriosclerosis, thrombosis, and vascular biology, 2020Am Heart Assoc
Objective: Lymphatics play an essential pathophysiological role in promoting fluid and
immune cell tissue clearance. Conversely, immune cells may influence lymphatic function
and remodeling. Recently, cardiac lymphangiogenesis has been proposed as a therapeutic
target to prevent heart failure after myocardial infarction (MI). We investigated the effects of
gene therapy to modulate cardiac lymphangiogenesis post-MI in rodents. Second, we
determined the impact of cardiac-infiltrating T cells on lymphatic remodeling in the heart …
Objective
Lymphatics play an essential pathophysiological role in promoting fluid and immune cell tissue clearance. Conversely, immune cells may influence lymphatic function and remodeling. Recently, cardiac lymphangiogenesis has been proposed as a therapeutic target to prevent heart failure after myocardial infarction (MI). We investigated the effects of gene therapy to modulate cardiac lymphangiogenesis post-MI in rodents. Second, we determined the impact of cardiac-infiltrating T cells on lymphatic remodeling in the heart.
Approach and Results
Comparing adenoviral versus adeno-associated viral gene delivery in mice, we found that only sustained VEGF (vascular endothelial growth factor)-CC156S therapy, achieved by adeno-associated viral vectors, increased cardiac lymphangiogenesis, and led to reduced cardiac inflammation and dysfunction by 3 weeks post-MI. Conversely, inhibition of VEGF-C/-D signaling, through adeno-associated viral delivery of soluble VEGFR3 (vascular endothelial growth factor receptor 3), limited infarct lymphangiogenesis. Unexpectedly, this treatment improved cardiac function post-MI in both mice and rats, linked to reduced infarct thinning due to acute suppression of T-cell infiltration. Finally, using pharmacological, genetic, and antibody-mediated prevention of cardiac T-cell recruitment in mice, we discovered that both CD4+ and CD8+ T cells potently suppress, in part through interferon-γ, cardiac lymphangiogenesis post-MI.
Conclusions
We show that resolution of cardiac inflammation after MI may be accelerated by therapeutic lymphangiogenesis based on adeno-associated viral gene delivery of VEGF-CC156S. Conversely, our work uncovers a major negative role of cardiac-recruited T cells on lymphatic remodeling. Our results give new insight into the interconnection between immune cells and lymphatics in orchestration of cardiac repair after injury.
Am Heart Assoc