Articles

< Previous         Next >  
Cardiac-specific knockout of ETA receptor mitigates low ambient temperature-induced cardiac hypertrophy and contractile dysfunction Free
Yingmei Zhang1,2,†, Linlin Li2,3,†, Yinan Hua2, Jennifer M. Nunn2, Feng Dong2, Masashi Yanagisawa4, and Jun Ren1,2,*
1Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
2Center for Cardiovascular Research and Alternative Medicine, University of Wyoming, Laramie, WY 82071, USA
3Department of Pharmacology, Xinjiang Medical University, Urumqi, Xinjiang 830011, P. R. China
4Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA *Correspondence to:Jun Ren, E-mail: jren@uwyo.edu
J Mol Cell Biol, Volume 4, Issue 2, April 2012, 97-107,  https://doi.org/10.1093/jmcb/mjs002
Keyword: low ambient temperature, myocardial function, ETA receptor, TRPV1, mitochondria
Cold exposure is associated with oxidative stress and cardiac dysfunction. The endothelin (ET) system, which plays a key role in myocardial homeostasis, may participate in cold exposure-induced cardiovascular dysfunction. This study was designed to examine the role of ET-1 in cold stress-induced cardiac geometric and contractile responses. Wild-type (WT) and ETA receptor knockout (ETAKO) mice were assigned to normal or cold exposure (4°C) environment for 2 and 5 weeks prior to evaluation of cardiac geometry, contractile, and intracellular Ca2+ properties. Levels of the temperature sensor transient receptor potential vanilloid (TRPV1), mitochondrial proteins for biogenesis and oxidative phosphorylation, including UCP2, HSP90, and PGC1α were evaluated. Cold stress triggered cardiac hypertrophy, depressed myocardial contractile capacity, including fractional shortening, peak shortening, and maximal velocity of shortening/relengthening, reduced intracellular Ca2+ release, prolonged intracellular Ca2+ decay and relengthening duration, generation of ROS and superoxide, as well as apoptosis, the effects of which were blunted by ETAKO. Western blotting revealed downregulated TRPV1 and PGC1α as well as upregulated UCP2 and activation of GSK3β, GATA4, and CREB in cold-stressed WT mouse hearts, which were obliterated by ETAKO. Levels of HSP90, an essential regulator for thermotolerance, were unchanged. The TRPV1 agonist SA13353 attenuated whereas TRPV1 antagonist capsazepine mimicked cold stress- or ET-1-induced cardiac anomalies. The GSK3β inhibitor SB216763 ablated cold stress-induced cardiac contractile (but not remodeling) changes and ET-1-induced TRPV1 downregulation. These data suggest that ETAKO protects against cold exposure-induced cardiac remodeling and dysfunction mediated through TRPV1 and mitochondrial function.