Unknown

Dataset Information

0

A novel, de novo mutation in the PRKAG2 gene: infantile-onset phenotype and the signaling pathway involved.


ABSTRACT: PRKAG2 encodes the ?2-subunit isoform of 5'-AMP-activated protein kinase (AMPK), a heterotrimeric enzyme with major roles in the regulation of energy metabolism in response to cellular stress. Mutations in PRKAG2 have been implicated in a unique hypertrophic cardiomyopathy (HCM) characterized by cardiac glycogen overload, ventricular preexcitation, and hypertrophy. We identified a novel, de novo PRKAG2 mutation (K475E) in a neonate with prenatal onset of HCM. We aimed to investigate the cellular impact, signaling pathways involved, and therapeutic options for K475E mutation using cells stably expressing human wild-type (WT) or the K475E mutant. In human embryonic kidney-293 cells, the K475E mutation induced a marked increase in the basal phosphorylation of T172 and AMPK activity, reduced sensitivity to AMP in allosteric activation, and a loss of response to phenformin. In H9c2 cardiomyocytes, the K475E mutation induced inhibition of AMPK and reduced the response to phenformin and increases in the phosphorylation of p70S6 kinase (p70S6K) and eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1). Primary fibroblasts from the patient with the K475E mutation also showed marked increases in the phosphorylation of p70S6K and 4E-BP1 compared with those from age-matched, nondiseased controls. Moreover, overexpression of K475E induced hypertrophy in H9c2 cells, which was effectively reversed by treatment with rapamycin. Taken together, we have identified a novel, de novo infantile-onset PRKAG2 mutation causing HCM. Our study suggests the K475E mutation induces alteration in basal AMPK activity and results in a hypertrophy phenotype involving the mechanistic target of rapamycin signaling pathway, which can be reversed with rapamycin.NEW & NOTEWORTHY We identified a novel, de novo PRKAG2 mutation (K475E) in the cystathionine ?-synthase 3 repeat, a region critical for AMP binding but with no previous reported mutation. Our data suggest the mutation affects AMP-activated protein kinase activity, activates cell growth pathways, and results in cardiac hypertrophy, which can be reversed with rapamycin.

SUBMITTER: Xu Y 

PROVIDER: S-EPMC5582920 | biostudies-literature | 2017 Aug

REPOSITORIES: biostudies-literature

altmetric image

Publications

A novel, de novo mutation in the <i>PRKAG2</i> gene: infantile-onset phenotype and the signaling pathway involved.

Xu Yanchun Y   Gray A A   Hardie D Grahame DG   Uzun Alper A   Shaw Sunil S   Padbury James J   Phornphutkul Chanika C   Tseng Yi-Tang YT  

American journal of physiology. Heart and circulatory physiology 20170526 2


<i>PRKAG2</i> encodes the γ<sub>2</sub>-subunit isoform of 5'-AMP-activated protein kinase (AMPK), a heterotrimeric enzyme with major roles in the regulation of energy metabolism in response to cellular stress. Mutations in <i>PRKAG2</i> have been implicated in a unique hypertrophic cardiomyopathy (HCM) characterized by cardiac glycogen overload, ventricular preexcitation, and hypertrophy. We identified a novel, de novo <i>PRKAG2</i> mutation (K475E) in a neonate with prenatal onset of HCM. We a  ...[more]

Similar Datasets

| S-EPMC3669303 | biostudies-literature
| S-EPMC6699192 | biostudies-literature
2019-09-02 | PXD011355 | Pride
| S-EPMC5002930 | biostudies-literature
| S-EPMC6122024 | biostudies-other
| S-EPMC3980418 | biostudies-literature
| S-EPMC5460159 | biostudies-literature
| S-EPMC6318765 | biostudies-literature
| S-EPMC5113095 | biostudies-literature
| S-EPMC8463406 | biostudies-literature