Format

Send to

Choose Destination
Proc Natl Acad Sci U S A. 2018 Sep 11;115(37):9276-9281. doi: 10.1073/pnas.1714538115. Epub 2018 Aug 27.

Telomere shortening is a hallmark of genetic cardiomyopathies.

Chang ACY1,2,3,4,5, Chang ACH6,2, Kirillova A6,2, Sasagawa K6,2, Su W6,2, Weber G6,2,3, Lin J7, Termglinchan V3, Karakikes I3,8, Seeger T3, Dainis AM3,9, Hinson JT10,11, Seidman J12, Seidman CE10,12,13, Day JW14, Ashley E3,4,5, Wu JC3,15, Blau HM1,2,3.

Author information

1
Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305; acychang@stanford.edu hblau@stanford.edu.
2
Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305.
3
Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305.
4
Stanford Center for Inherited Cardiovascular Disease, Stanford University School of Medicine, Stanford, CA 94305.
5
Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305.
6
Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305.
7
Department of Biochemistry and Biophysics, School of Medicine, University of California, San Francisco, CA 94158.
8
Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA 94305.
9
Department of Genetics, Stanford University, Stanford, CA 94305.
10
Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA 02115.
11
Cardiology Division, UConn Health and The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032.
12
Department of Genetics, Harvard Medical School, Boston, MA 02115.
13
Howard Hughes Medical Institute, Chevy Chase, MD 20815.
14
Department of Neurology, Stanford University, Stanford, CA 94304.
15
Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA 94305.

Abstract

This study demonstrates that significantly shortened telomeres are a hallmark of cardiomyocytes (CMs) from individuals with end-stage hypertrophic cardiomyopathy (HCM) or dilated cardiomyopathy (DCM) as a result of heritable defects in cardiac proteins critical to contractile function. Positioned at the ends of chromosomes, telomeres are DNA repeats that serve as protective caps that shorten with each cell division, a marker of aging. CMs are a known exception in which telomeres remain relatively stable throughout life in healthy individuals. We found that, relative to healthy controls, telomeres are significantly shorter in CMs of genetic HCM and DCM patient tissues harboring pathogenic mutations: TNNI3, MYBPC3, MYH7, DMD, TNNT2, and TTN Quantitative FISH (Q-FISH) of single cells revealed that telomeres were significantly reduced by 26% in HCM and 40% in DCM patient CMs in fixed tissue sections compared with CMs from age- and sex-matched healthy controls. In the cardiac tissues of the same patients, telomere shortening was not evident in vascular smooth muscle cells that do not express or require the contractile proteins, an important control. Telomere shortening was recapitulated in DCM and HCM CMs differentiated from patient-derived human-induced pluripotent stem cells (hiPSCs) measured by two independent assays. This study reveals telomere shortening as a hallmark of genetic HCM and DCM and demonstrates that this shortening can be modeled in vitro by using the hiPSC platform, enabling drug discovery.

KEYWORDS:

dilated cardiomyopathy; hiPSC-CM; hypertrophy cardiomyopathy; telomere

PMID:
30150400
PMCID:
PMC6140486
DOI:
10.1073/pnas.1714538115
[Indexed for MEDLINE]
Free PMC Article

Conflict of interest statement

Conflict of interest statement: The sponsor declares a conflict of interest. J.L. is a cofounder and consultant to Telomere Diagnostics. The company played no role in this research.

Supplemental Content

Full text links

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