Endoplasmic reticulum protein TXNDC5 augments myocardial fibrosis by facilitating extracellular matrix protein folding and redox-sensitive cardiac fibroblast …

YC Shih, CL Chen, Y Zhang, RL Mellor… - Circulation …, 2018 - Am Heart Assoc
YC Shih, CL Chen, Y Zhang, RL Mellor, EM Kanter, Y Fang, HC Wang, CT Hung, JY Nong…
Circulation research, 2018Am Heart Assoc
Rationale: Cardiac fibrosis plays a critical role in the pathogenesis of heart failure. Excessive
accumulation of extracellular matrix (ECM) resulting from cardiac fibrosis impairs cardiac
contractile function and increases arrhythmogenicity. Current treatment options for cardiac
fibrosis, however, are limited, and there is a clear need to identify novel mediators of cardiac
fibrosis to facilitate the development of better therapeutics. Exploiting coexpression gene
network analysis on RNA sequencing data from failing human heart, we identified TXNDC5 …
Rationale:
Cardiac fibrosis plays a critical role in the pathogenesis of heart failure. Excessive accumulation of extracellular matrix (ECM) resulting from cardiac fibrosis impairs cardiac contractile function and increases arrhythmogenicity. Current treatment options for cardiac fibrosis, however, are limited, and there is a clear need to identify novel mediators of cardiac fibrosis to facilitate the development of better therapeutics. Exploiting coexpression gene network analysis on RNA sequencing data from failing human heart, we identified TXNDC5 (thioredoxin domain containing 5), a cardiac fibroblast (CF)–enriched endoplasmic reticulum protein, as a potential novel mediator of cardiac fibrosis, and we completed experiments to test this hypothesis directly.
Objective:
The objective of this study was to determine the functional role of TXNDC5 in the pathogenesis of cardiac fibrosis.
Methods and Results:
RNA sequencing and Western blot analyses revealed that TXNDC5 mRNA and protein were highly upregulated in failing human left ventricles and in hypertrophied/failing mouse left ventricle. In addition, cardiac TXNDC5 mRNA expression levels were positively correlated with those of transcripts encoding transforming growth factor β1 and ECM proteins in vivo. TXNDC5 mRNA and protein were increased in human CF (hCF) under transforming growth factor β1 stimulation in vitro. Knockdown of TXNDC5 attenuated transforming growth factor β1–induced hCF activation and ECM protein upregulation independent of SMAD3 (SMAD family member 3), whereas increasing expression of TXNDC5 triggered hCF activation and proliferation and increased ECM protein production. Further experiments showed that TXNDC5, a protein disulfide isomerase, facilitated ECM protein folding and that depletion of TXNDC5 led to ECM protein misfolding and degradation in CF. In addition, TXNDC5 promotes hCF activation and proliferation by enhancing c-Jun N-terminal kinase activity via increased reactive oxygen species, derived from NAD(P)H oxidase 4. Transforming growth factor β1–induced TXNDC5 upregulation in hCF was dependent on endoplasmic reticulum stress and activating transcription factor 6–mediated transcriptional control. Targeted disruption of Txndc5 in mice (Txndc5−/−) revealed protective effects against isoproterenol-induced cardiac hypertrophy, reduced fibrosis (by ≈70%), and markedly improved left ventricle function; post-isoproterenol left ventricular ejection fraction was 59.1±1.5 versus 40.1±2.5 (P<0.001) in Txndc5−/− versus wild-type mice, respectively.
Conclusions:
The endoplasmic reticulum protein TXNDC5 promotes cardiac fibrosis by facilitating ECM protein folding and CF activation via redox-sensitive c-Jun N-terminal kinase signaling. Loss of TXNDC5 protects against β agonist–induced cardiac fibrosis and contractile dysfunction. Targeting TXNDC5, therefore, could be a powerful new therapeutic approach to mitigate excessive cardiac fibrosis, thereby improving cardiac function and outcomes in patients with heart failure.
Am Heart Assoc