Abstract

Introduction: Targeted-ablation of guanylyl cyclase/natriuretic peptide receptor-A (GC-A/NPRA) gene ( Npr1 ) in mice exhibits hypertension and provokes congestive heart failure; however, the underlying mechanisms are not well understood. Hypothesis: To determine whether transforming growth factor-beta 1 (TGF)-β1 receptor antagonist, GW788388 inhibits the development of cardiac fibrosis and remodeling in GC-A/NPRA gene-disrupted mice. Methods: The adult male (16-20 weeks) Npr1 null mutant ( Npr1 -/- , 0-copy), heterozygous ( Npr1 +/- , 1-copy), and wild-type ( Npr1 +/+ , 2-copy) mice were orally administered with TGF-β1 receptor antagonist, GW788388 (2 mg/kg/day) for 28 days. The expression of cardiac fibrotic markers was analyzed using real-time PCR and Western blot. Heart weight-to-body weight (HW/BW) ratios were determined and heart functions were measured by echocardiographic analysis. Results: The Npr1 -/- mice showed markedly increased cardiac fibrosis and HW/BW ratio with increased expression of collagen-1α (3.5-fold), monocyte chemoattractant protein (4-fold), connective tissue growth factor (CTGF, 5-fold), α-smooth muscle actin (α-SMA, 4-fold), TGF-βRI (4-fold), TGF-βRII (3.5-fold) and SMAD proteins (SMAD-2, 5-fold; SMAD-3, 3-fold) compared with Npr1 +/+ mice. The expression of phosphorylated extracellular-regulated kinase (pERK1/2) was also up-regulated in Npr1 -/- mice. The treatment of Npr1 -/- mice with GW788388 significantly prevented the development of cardiac fibrosis and down-regulated the expression of fibrotic markers and SMAD proteins compared to vehicle-treated mice. In contrast, the expression of pERK1/2 proteins was unaffected in GW7885388-treated mice excluding the involvement of non-genomic pathway. The left ventricular dimensions (systole and diastole) and fractional shortening were significantly improved in the drug-treated Npr1 -/- mice. Conclusions: The results suggest that the cardiac fibrosis, remodeling, and dysfunction in Npr1 -/- mice are regulated through TGF-β-mediated SMAD-dependent canonical pathway. The findings will be important for the development of new molecular therapeutic targets for the treatment of cardiac fibrosis, remodeling, and dysfunction in human patients.

Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.