LAUSR.org creates dashboard-style pages of related content for over 1.5 million academic articles. Sign Up to like articles & get recommendations!

AMPK&agr;2 Protects Against the Development of Heart Failure by Enhancing Mitophagy via PINK1 Phosphorylation

Photo by freestocks from unsplash

Rationale: Mitochondrial dysfunction plays an important role in heart failure (HF). However, the molecular mechanisms regulating mitochondrial functions via selective mitochondrial autophagy (mitophagy) are poorly understood. Objective: We sought to… Click to show full abstract

Rationale: Mitochondrial dysfunction plays an important role in heart failure (HF). However, the molecular mechanisms regulating mitochondrial functions via selective mitochondrial autophagy (mitophagy) are poorly understood. Objective: We sought to determine the role of AMPK (AMP-activated protein kinase) in selective mitophagy during HF. Methods and Results: An isoform shift from AMPK&agr;2 to AMPK&agr;1 was observed in failing heart samples from HF patients and transverse aortic constriction–induced mice, accompanied by decreased mitophagy and mitochondrial function. The recombinant adeno-associated virus Serotype 9-mediated overexpression of AMPK&agr;2 in mouse hearts prevented the development of transverse aortic constriction–induced chronic HF by increasing mitophagy and improving mitochondrial function. In contrast, AMPK&agr;2−/− mutant mice exhibited an exacerbation of the early progression of transverse aortic constriction–induced HF via decreases in cardiac mitophagy. In isolated adult mouse cardiomyocytes, AMPK&agr;2 overexpression mechanistically rescued the impairment of mitophagy after phenylephrine stimulation for 24 hours. Genetic knockdown of AMPK&agr;2, but not AMPK&agr;1, by short interfering RNA suppressed the early phase (6 hours) of phenylephrine-induced compensatory increases in mitophagy. Furthermore, AMPK&agr;2 specifically interacted with phosphorylated PINK1 (PTEN-induced putative kinase 1) at Ser495 after phenylephrine stimulation. Subsequently, phosphorylated PINK1 recruited the E3 ubiquitin ligase, Parkin, to depolarized mitochondria, and then enhanced the role of the PINK1–Parkin–SQSTM1 (sequestosome-1) pathway involved in cardiac mitophagy. This increase in cardiac mitophagy was accompanied by the elimination of damaged mitochondria, improvement in mitochondrial function, decrease in reactive oxygen species production, and apoptosis of cardiomyocytes. Finally, Ala mutation of PINK1 at Ser495 partially suppressed AMPK&agr;2 overexpression-induced mitophagy and improvement of mitochondrial function in phenylephrine-stimulated cardiomyocytes, whereas Asp (phosphorylation mimic) mutation promoted mitophagy after phenylephrine stimulation. Conclusions: In failing hearts, the dominant AMPK&agr; isoform switched from AMPK&agr;2 to AMPK&agr;1, which accelerated HF. The results show that phosphorylation of Ser495 in PINK1 by AMPK&agr;2 was essential for efficient mitophagy to prevent the progression of HF.

Keywords: heart; ampk agr; ampk; pink1; mitophagy

Journal Title: Circulation Research
Year Published: 2018

Link to full text (if available)


Share on Social Media:                               Sign Up to like & get
recommendations!

Related content

More Information              News              Social Media              Video              Recommended



                Click one of the above tabs to view related content.