Abstract

Diastolic dysfunction is a prominent feature of cardiac aging in both mice and humans. We show here that 8-week treatment of old mice with the mitochondrial targeted peptide SS-31 (elamipretide) can substantially reverse this deficit. SS-31 normalized the increase in proton leak and reduced mitochondrial ROS in cardiomyocytes from old mice, accompanied by reduced protein oxidation and a shift towards a more reduced protein thiol redox state in old hearts. Improved diastolic function was concordant with increased phosphorylation of cMyBP-C Ser282 but was independent of titin isoform shift. Late-life viral expression of mitochondrial-targeted catalase (mCAT) produced similar functional benefits in old mice and SS-31 did not improve cardiac function of old mCAT mice, implicating normalizing mitochondrial oxidative stress as an overlapping mechanism. These results demonstrate that pre-existing cardiac aging phenotypes can be reversed by targeting mitochondrial dysfunction and implicate mitochondrial energetics and redox signaling as therapeutic targets for cardiac aging.

Highlights

  • Mitochondrial dysfunction is one of the hallmarks of aging (Lopez-Otın et al, 2013)

  • The main findings of the study are: 1) enhancing mitochondrial function at late-life by administration of mitochondrial-targeted SS-31 peptide or AAV-mediated expression of mitochondrial targeted catalase can reverse pre-existing cardiac dysfunction in old mice; 2) SS-31 treatment normalizes the age-related increase in mitochondrial proton leak, reduces reactive oxygen species (ROS) production by old cardiomyocytes, and reduces protein oxidative modifications; 3) the rescue of diastolic function by SS-31 in old mice is due, at least in part, to reversal of hypo-phosphorylation of myofilament protein cardiac myosin binding protein C (cMyBP-C); and 4) SS-31 treatment and mitochondrial-targeted catalase (mCAT) expression, while similar in many ways, differentially regulate myofilament protein phosphorylation

  • These findings are summarized in a proposed mechanistic model of how SS-31 treatment and mCAT expression improve mitochondrial function and regulates myofilament properties to improve cardiomyocytes relaxation and reversing age-related cardiac dysfunction (Figure 8)

Read more

Summary

Introduction

Mitochondrial dysfunction is one of the hallmarks of aging (Lopez-Otın et al, 2013). While mitochondria generate the bulk of cellular ATP, they are the major source of reactive oxygen species (ROS) in most cells. SS-31 treatment was previously shown to reduce mitochondrial oxidative damage and prevent pressure overload-induced cardiac hypertrophy and failure in a manner that was highly similar to mCAT (Dai et al, 2011b; Dai et al, 2013; Dai et al, 2012) While these and other studies have shown that combating mitochondrial ROS during the course of a lifetime or during work and pressure overload stress can prevent mitochondrial dysfunction and attenuate cardiac functional decline (Dai et al, 2014a; Dai et al, 2017), it has not been established whether delivering such interventions in later life can rescue pre-existing mitochondrial and cardiac dysfunction. We demonstrate that mitochondrial-targeted interventions can improve mitochondrial function and reverse pre-existing cardiac dysfunction in old mice

Results
Discussion
Findings
Materials and methods
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