4.7 Article

Methamphetamine causes cardiovascular dysfunction via cystathionine gamma lyase and hydrogen sulfide depletion

Journal

REDOX BIOLOGY
Volume 57, Issue -, Pages -

Publisher

ELSEVIER
DOI: 10.1016/j.redox.2022.102480

Keywords

Cystathionine gamma lyase; Hydrogen sulfide; Nitric oxide; Methamphetamine; Endothelial dysfunction; Cardiomyopathy

Funding

  1. Institutional Development Award (IDeA) from the National Institutes of General Medical Sciences of the NIH [P20GM121307, HL149264]
  2. NIH [HL098435, HL133497, HL141155, HL131844, HL122354, HL145753]
  3. NIH COBRE Pilot Grant

Ask authors/readers for more resources

Methamphetamine use has been found to significantly decrease cardiovascular function by reducing the availability of cystathionine gamma lyase (CSE), hydrogen sulfide (H2S), and nitric oxide (NO). This leads to endothelial dysfunction and pathological changes in the heart. Increasing CSE/H2S bioavailability may be a potential therapeutic intervention for attenuating METH-induced cardiovascular disease.
Methamphetamine (METH) is an addictive illicit drug used worldwide that causes significant damage to blood vessels resulting in cardiovascular dysfunction. Recent studies highlight increased prevalence of cardiovascular disease (CVD) and associated complications including hypertension, vasospasm, left ventricular hypertrophy, and coronary artery disease in younger populations due to METH use. Here we report that METH administration in a mouse model of 'binge and crash' decreases cardiovascular function via cystathionine gamma lyase (CSE), hydrogen sulfide (H2S), nitric oxide (NO) (CSE/H2S/NO) dependent pathway. METH significantly reduced H2S and NO bioavailability in plasma and skeletal muscle tissues co-incident with a significant reduction in flow-mediated vasodilation (FMD) and blood flow velocity revealing endothelial dysfunction. METH administration also reduced cardiac ejection fraction (EF) and fractional shortening (FS) associated with increased tissue and perivascular fibrosis. Importantly, METH treatment selectively decreased CSE expression and sulfide bioavail-ability along with reduced eNOS phosphorylation and NO levels. Exogenous sulfide therapy or endothelial CSE transgenic overexpression corrected cardiovascular and associated pathological responses due to METH impli-cating a central molecular regulatory pathway for tissue pathology. These findings reveal that therapeutic intervention targeting CSE/H2S bioavailability may be useful in attenuating METH mediated cardiovascular disease.

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