Abstract

Renin is the rate‐limiting enzyme of the renin–angiotensin system cascade, which drives the pathophysiological progression of heart failure. Species differences in the amino acid sequence of the catalytic domain of renin limit evaluations of the potency and efficacy of human renin inhibitors in animal models, and a high dose of inhibitors is usually needed to show its organ‐protective effects in rodents. In the present study, we developed a novel murine heart failure model (triple‐tg) to enable us to evaluate the cardioprotective effect of renin inhibitors at more relevant doses for humans, by cross‐breeding calsequestrin transgenic (CSQ‐tg) mice with human renin and human angiotensinogen double‐transgenic mice. The triple‐tg mice exhibited increased plasma renin activity, worsened cardiac hypertrophy, and higher mortality compared to CSQ‐tg mice. Triple‐tg mice treated with 10 mg·kg−1 of TAK‐272 (imarikiren/SCO‐272), an orally active direct renin inhibitor, exhibited improvements in heart failure phenotypes, such as cardiac hypertrophy and survival rate; however, a dose of 300 mg·kg−1 was required to improve symptoms in CSQ‐tg mice. Our results suggest that this newly generated triple‐tg heart failure model is useful to evaluate the cardioprotective effects of human renin inhibitors at clinically relevant doses, thereby minimizing the concerns of off‐target effects related to much higher drug exposure than that achieved in clinical study.

Highlights

  • Renin is the rate-limiting enzyme of the renin–angiotensin system cascade, which drives the pathophysiological progression of heart failure

  • We developed a novel murine heart failure model to enable us to evaluate the cardioprotective effect of renin inhibitors at more relevant doses for humans, by cross-breeding calsequestrin transgenic (CSQ-tg) mice with human renin and human angiotensinogen double-transgenic mice

  • Our results suggest that this newly generated triple-tg heart failure model is useful to evaluate the cardioprotective effects of human renin inhibitors at clinically relevant doses, thereby minimizing the concerns of off-target effects related to much higher drug exposure than that achieved in clinical study

Read more

Summary

Materials and methods

The transgenic DBA/2N mice with cardiac-specific overexpression of canine CSQ were originally developed in a facility at the Indiana University School of Medicine [11] and bred by Takeda Rabics (Osaka, Japan). 930 bp) or canine CSQ primers (CTCTGACAGAGAAGCAGGCACTTTACATGG and GATGAACAGGTGTGTTCTCTTCAT, 407 bp) to confirm the expression of these genes; all the mice were distinguished as triple-tg, CSQ-tg, RA-tg, or WT mice based on the genotypes As both male and female mice showed comparable symptoms and survival rates in the preliminary studies, both sexes were used in this study due to limited animal availability. Characterization of cardiovascular parameters and PRA on transgenic mice The systolic blood pressure (SBP) of male WT, RA-tg, CSQ-tg, and triple-tg mice (n = 10 for each group) was measured by the tail-cuff method using a BP-98A blood pressure analyzer (Softron, Tokyo, Japan) under blind conditions at 4 weeks old. Twenty triple-tg mice (male: 8; female: 12) were treated with vehicle or TAK-272 once daily starting at 4 weeks of age for 24 days. The Kaplan–Meier method with log-rank test was used for survival analysis

Results
Discussion
Conflict of interest
Full Text
Published version (Free)

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