4.7 Article

AMPK2 Protects Against the Development of Heart Failure by Enhancing Mitophagy via PINK1 Phosphorylation

期刊

CIRCULATION RESEARCH
卷 122, 期 5, 页码 712-729

出版社

LIPPINCOTT WILLIAMS & WILKINS
DOI: 10.1161/CIRCRESAHA.117.312317

关键词

autophagy; constriction; heart failure; mice; mitochondria

资金

  1. National Natural Science Foundation of China [81630010, 91439203, 31571197]
  2. National Basic Research Program of China [2012CB518004]
  3. Fundamental Research Funds for the Central Universities [2015ZDTD044]
  4. National Institutes of Health [HL079584, HL080499, HL089920, HL110488, CA213022, AG047776, HL128014, HL132500, HL137371]
  5. NATIONAL CANCER INSTITUTE [R01CA213022] Funding Source: NIH RePORTER
  6. NATIONAL HEART, LUNG, AND BLOOD INSTITUTE [R01HL128014, R01HL080499, R01HL140954, R01HL137371, R01HL089920, R01HL110488, R01HL079584, R01HL074399, R01HL132500] Funding Source: NIH RePORTER
  7. NATIONAL INSTITUTE ON AGING [R01AG047776] Funding Source: NIH RePORTER

向作者/读者索取更多资源

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 AMPK2 to AMPK1 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 AMPK2 in mouse hearts prevented the development of transverse aortic constriction-induced chronic HF by increasing mitophagy and improving mitochondrial function. In contrast, AMPK2(-/-) 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, AMPK2 overexpression mechanistically rescued the impairment of mitophagy after phenylephrine stimulation for 24 hours. Genetic knockdown of AMPK2, but not AMPK1, by short interfering RNA suppressed the early phase (6 hours) of phenylephrine-induced compensatory increases in mitophagy. Furthermore, AMPK2 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 AMPK2 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 isoform switched from AMPK2 to AMPK1, which accelerated HF. The results show that phosphorylation of Ser495 in PINK1 by AMPK2 was essential for efficient mitophagy to prevent the progression of HF.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据