4.7 Article

Mitochondrial H2S Regulates BCAA Catabolism in Heart Failure

Journal

CIRCULATION RESEARCH
Volume 131, Issue 3, Pages 222-235

Publisher

LIPPINCOTT WILLIAMS & WILKINS
DOI: 10.1161/CIRCRESAHA.121.319817

Keywords

amino acids; branched-chain; cell death; heart failure; hydrogen sulfide; mitochondrial respiration

Funding

  1. NIH National Heart, Lung, and Blood Institute [R01 HL146098, R01 HL146514, R01 HL137711, R01 HL151398, R01 HL133167, R35 HL155651]
  2. American Heart Association Postdoctoral Grant [20POST3520075, 18POST34020143]

Ask authors/readers for more resources

This study reveals that the reduction of 3-MST in the myocardium is associated with heart failure. Knockout of 3-MST worsens cardiac and vascular dysfunction in a mouse model of pressure overload-induced heart failure. Restoring branched-chain amino acid metabolism and administrating an H2S donor can alleviate the detrimental effects of 3-MST deficiency in heart failure.
Background: Hydrogen sulfide (H2S) exerts mitochondria-specific actions that include the preservation of oxidative phosphorylation, biogenesis, and ATP synthesis, while inhibiting cell death. 3-MST (3-mercaptopyruvate sulfurtransferase) is a mitochondrial H2S-producing enzyme whose functions in the cardiovascular disease are not fully understood. In the current study, we investigated the effects of global 3-MST deficiency in the setting of pressure overload-induced heart failure. Methods: Human myocardial samples obtained from patients with heart failure undergoing cardiac surgeries were probed for 3-MST protein expression. 3-MST knockout mice and C57BL/6J wild-type mice were subjected to transverse aortic constriction to induce pressure overload heart failure with reduced ejection fraction. Cardiac structure and function, vascular reactivity, exercise performance, mitochondrial respiration, and ATP synthesis efficiency were assessed. In addition, untargeted metabolomics were utilized to identify key pathways altered by 3-MST deficiency. Results: Myocardial 3-MST was significantly reduced in patients with heart failure compared with nonfailing controls. 3-MST KO mice exhibited increased accumulation of branched-chain amino acids in the myocardium, which was associated with reduced mitochondrial respiration and ATP synthesis, exacerbated cardiac and vascular dysfunction, and worsened exercise performance following transverse aortic constriction. Restoring myocardial branched-chain amino acid catabolism with 3,6-dichlorobenzo1[b]thiophene-2-carboxylic acid (BT2) and administration of a potent H2S donor JK-1 ameliorates the detrimental effects of 3-MST deficiency in heart failure with reduced ejection fraction. Conclusions: Our data suggest that 3-MST derived mitochondrial H2S may play a regulatory role in branched-chain amino acid catabolism and mediate critical cardiovascular protection in heart failure.

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