4.7 Article

Stearoyl-CoA Desaturase Regulates Angiogenesis and Energy Metabolism in Ischemic Cardiomyocytes

Journal

Publisher

MDPI
DOI: 10.3390/ijms231810459

Keywords

heart; cytokines; hypoxia; fatty acids; substrate utilization

Funding

  1. National Science Center, Poland [UMO-2016/22/E/NZ4/00650]

Ask authors/readers for more resources

New blood vessel formation is a crucial component of cardiac repair after myocardial infarction (MI). This study identifies stearoyl-CoA desaturase (SCD) as a significant regulator of angiogenesis in the hypoxic myocardium, independent of HIF1 alpha expression. Inhibition of SCD activity disrupts the expression and secretion of proangiogenic factors and impairs the proangiogenic potential of ischemic cardiomyocytes. Additionally, SCD is involved in shaping the energy metabolism of the ischemic heart. These findings highlight the role of SCD in cardiac repair processes after MI.
New blood vessel formation is a key component of the cardiac repair process after myocardial infarction (MI). Hypoxia following MI is a major driver of angiogenesis in the myocardium. Hypoxia-inducible factor 1 alpha (HIF1 alpha) is the key regulator of proangiogenic signaling. The present study found that stearoyl-CoA desaturase (SCD) significantly contributed to the induction of angiogenesis in the hypoxic myocardium independently of HIF1 alpha expression. The pharmacological inhibition of SCD activity in HL-1 cardiomyocytes and SCD knockout in an animal model disturbed the expression and secretion of proangiogenic factors including vascular endothelial growth factor-A, proinflammatory cytokines (interleukin-1 beta, interleukin-6, tumor necrosis factor alpha, monocyte chemoattractant protein-1, and Rantes), metalloproteinase-9, and platelet-derived growth factor in ischemic cardiomyocytes. These disturbances affected the proangiogenic potential of ischemic cardiomyocytes after SCD depletion. Together with the most abundant SCD1 isoform, the heart-specific SCD4 isoform emerged as an important regulator of new blood vessel formation in the murine post-MI myocardium. We also provide evidence that SCD shapes energy metabolism of the ischemic heart by maintaining the shift from fatty acids to glucose as the substrate that is used for adenosine triphosphate production. Furthermore, we propose that the regulation of the proangiogenic properties of hypoxic cardiomyocytes by key modulators of metabolic signaling such as adenosine monophosphate kinase, protein kinase B (AKT), and peroxisome-proliferator-activated receptor-gamma coactivator 1 alpha/peroxisome proliferator-activated receptor alpha depends on SCD to some extent. Thus, our results reveal a novel mechanism that links SCD to cardiac repair processes after MI.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available