Format

Send to

Choose Destination
Proc Natl Acad Sci U S A. 2016 Nov 15;113(46):13120-13125. Epub 2016 Oct 31.

Telomere shortening and metabolic compromise underlie dystrophic cardiomyopathy.

Chang AC1,2,3,4, Ong SG4,5, LaGory EL6, Kraft PE1,2,3, Giaccia AJ6, Wu JC4,5, Blau HM7,2,3,4.

Author information

1
Baxter Laboratory for Stem Cell Biology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305.
2
Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305.
3
Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford University, Stanford, CA 94305.
4
Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford University, Stanford, CA 94305.
5
Division of Cardiology, Department of Medicine Stanford and Department of Radiology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305.
6
Division of Radiation and Cancer Biology and Center for Clinical Sciences Research, Department of Radiation Oncology, Stanford University, Stanford, CA 94305.
7
Baxter Laboratory for Stem Cell Biology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305; hblau@stanford.edu.

Abstract

Duchenne muscular dystrophy (DMD) is an incurable X-linked genetic disease that is caused by a mutation in the dystrophin gene and affects one in every 3,600 boys. We previously showed that long telomeres protect mice from the lethal cardiac disease seen in humans with the same genetic defect, dystrophin deficiency. By generating the mdx4cv/mTRG2 mouse model with "humanized" telomere lengths, the devastating dilated cardiomyopathy phenotype seen in patients with DMD was recapitulated. Here, we analyze the degenerative sequelae that culminate in heart failure and death in this mouse model. We report progressive telomere shortening in developing mouse cardiomyocytes after postnatal week 1, a time when the cells are no longer dividing. This proliferation-independent telomere shortening is accompanied by an induction of a DNA damage response, evident by p53 activation and increased expression of its target gene p21 in isolated cardiomyocytes. The consequent repression of Pgc1α/β leads to impaired mitochondrial biogenesis, which, in conjunction with the high demands of contraction, leads to increased oxidative stress and decreased mitochondrial membrane potential. As a result, cardiomyocyte respiration and ATP output are severely compromised. Importantly, treatment with a mitochondrial-specific antioxidant before the onset of cardiac dysfunction rescues the metabolic defects. These findings provide evidence for a link between short telomere length and metabolic compromise in the etiology of dilated cardiomyopathy in DMD and identify a window of opportunity for preventive interventions.

KEYWORDS:

Duchenne muscular dystrophy; dilated cardiomyopathy; metabolic compromise; mitochondrial dysfunction; telomere

PMID:
27799523
PMCID:
PMC5135315
DOI:
10.1073/pnas.1615340113
[Indexed for MEDLINE]
Free PMC Article

Supplemental Content

Full text links

Icon for HighWire Icon for PubMed Central
Loading ...
Support Center