An Important Role for DNMT3A-Mediated DNA Methylation in Cardiomyocyte Metabolism and Contractility

Standard

An Important Role for DNMT3A-Mediated DNA Methylation in Cardiomyocyte Metabolism and Contractility. / Madsen, Alexandra; Höppner, Grit; Krause, Julia; Hirt, Marc N; Laufer, Sandra D; Schweizer, Michaela; Tan, Wilson Lek Wen; Mosqueira, Diogo; Anene-Nzelu, Chukwuemeka George; Lim, Ives; Foo, Roger S Y; Eschenhagen, Thomas; Stenzig, Justus.

in: CIRCULATION, Jahrgang 142, Nr. 16, 20.10.2020, S. 1562-1578.

Publikationen: SCORING: Beitrag in Fachzeitschrift/ZeitungSCORING: ZeitschriftenaufsatzForschungBegutachtung

Harvard

Madsen, A, Höppner, G, Krause, J, Hirt, MN, Laufer, SD, Schweizer, M, Tan, WLW, Mosqueira, D, Anene-Nzelu, CG, Lim, I, Foo, RSY, Eschenhagen, T & Stenzig, J 2020, 'An Important Role for DNMT3A-Mediated DNA Methylation in Cardiomyocyte Metabolism and Contractility', CIRCULATION, Jg. 142, Nr. 16, S. 1562-1578. https://doi.org/10.1161/CIRCULATIONAHA.119.044444

APA

Madsen, A., Höppner, G., Krause, J., Hirt, M. N., Laufer, S. D., Schweizer, M., Tan, W. L. W., Mosqueira, D., Anene-Nzelu, C. G., Lim, I., Foo, R. S. Y., Eschenhagen, T., & Stenzig, J. (2020). An Important Role for DNMT3A-Mediated DNA Methylation in Cardiomyocyte Metabolism and Contractility. CIRCULATION, 142(16), 1562-1578. https://doi.org/10.1161/CIRCULATIONAHA.119.044444

Vancouver

Bibtex

@article{8148483a2aad467b85a8c215ca055979,
title = "An Important Role for DNMT3A-Mediated DNA Methylation in Cardiomyocyte Metabolism and Contractility",
abstract = "BACKGROUND: DNA methylation acts as a mechanism of gene transcription regulation. It has recently gained attention as a possible therapeutic target in cardiac hypertrophy and heart failure. However, its exact role in cardiomyocytes remains controversial. Thus, we knocked out the main de novo DNA methyltransferase in cardiomyocytes, DNMT3A, in human induced pluripotent stem cells. Functional consequences of DNA methylation-deficiency under control and stress conditions were then assessed in human engineered heart tissue from knockout human induced pluripotent stem cell-derived cardiomyocytes.METHODS: DNMT3A was knocked out in human induced pluripotent stem cells by CRISPR/Cas9gene editing. Fibrin-based engineered heart tissue was generated from knockout and control human induced pluripotent stem cell-derived cardiomyocytes. Development and baseline contractility were analyzed by video-optical recording. Engineered heart tissue was subjected to different stress protocols, including serum starvation, serum variation, and restrictive feeding. Molecular, histological, and ultrastructural analyses were performed afterward.RESULTS: Knockout of DNMT3A in human cardiomyocytes had three main consequences for cardiomyocyte morphology and function: (1) Gene expression changes of contractile proteins such as higher atrial gene expression and lower MYH7/MYH6 ratio correlated with different contraction kinetics in knockout versus wild-type; (2) Aberrant activation of the glucose/lipid metabolism regulator peroxisome proliferator-activated receptor gamma was associated with accumulation of lipid vacuoles within knockout cardiomyocytes; (3) Hypoxia-inducible factor 1α protein instability was associated with impaired glucose metabolism and lower glycolytic enzyme expression, rendering knockout-engineered heart tissue sensitive to metabolic stress such as serum withdrawal and restrictive feeding.CONCLUSION: The results suggest an important role of DNA methylation in the normal homeostasis of cardiomyocytes and during cardiac stress, which could make it an interesting target for cardiac therapy.",
author = "Alexandra Madsen and Grit H{\"o}ppner and Julia Krause and Hirt, {Marc N} and Laufer, {Sandra D} and Michaela Schweizer and Tan, {Wilson Lek Wen} and Diogo Mosqueira and Anene-Nzelu, {Chukwuemeka George} and Ives Lim and Foo, {Roger S Y} and Thomas Eschenhagen and Justus Stenzig",
year = "2020",
month = oct,
day = "20",
doi = "10.1161/CIRCULATIONAHA.119.044444",
language = "English",
volume = "142",
pages = "1562--1578",
journal = "CIRCULATION",
issn = "0009-7322",
publisher = "Lippincott Williams and Wilkins",
number = "16",

}

RIS

TY - JOUR

T1 - An Important Role for DNMT3A-Mediated DNA Methylation in Cardiomyocyte Metabolism and Contractility

AU - Madsen, Alexandra

AU - Höppner, Grit

AU - Krause, Julia

AU - Hirt, Marc N

AU - Laufer, Sandra D

AU - Schweizer, Michaela

AU - Tan, Wilson Lek Wen

AU - Mosqueira, Diogo

AU - Anene-Nzelu, Chukwuemeka George

AU - Lim, Ives

AU - Foo, Roger S Y

AU - Eschenhagen, Thomas

AU - Stenzig, Justus

PY - 2020/10/20

Y1 - 2020/10/20

N2 - BACKGROUND: DNA methylation acts as a mechanism of gene transcription regulation. It has recently gained attention as a possible therapeutic target in cardiac hypertrophy and heart failure. However, its exact role in cardiomyocytes remains controversial. Thus, we knocked out the main de novo DNA methyltransferase in cardiomyocytes, DNMT3A, in human induced pluripotent stem cells. Functional consequences of DNA methylation-deficiency under control and stress conditions were then assessed in human engineered heart tissue from knockout human induced pluripotent stem cell-derived cardiomyocytes.METHODS: DNMT3A was knocked out in human induced pluripotent stem cells by CRISPR/Cas9gene editing. Fibrin-based engineered heart tissue was generated from knockout and control human induced pluripotent stem cell-derived cardiomyocytes. Development and baseline contractility were analyzed by video-optical recording. Engineered heart tissue was subjected to different stress protocols, including serum starvation, serum variation, and restrictive feeding. Molecular, histological, and ultrastructural analyses were performed afterward.RESULTS: Knockout of DNMT3A in human cardiomyocytes had three main consequences for cardiomyocyte morphology and function: (1) Gene expression changes of contractile proteins such as higher atrial gene expression and lower MYH7/MYH6 ratio correlated with different contraction kinetics in knockout versus wild-type; (2) Aberrant activation of the glucose/lipid metabolism regulator peroxisome proliferator-activated receptor gamma was associated with accumulation of lipid vacuoles within knockout cardiomyocytes; (3) Hypoxia-inducible factor 1α protein instability was associated with impaired glucose metabolism and lower glycolytic enzyme expression, rendering knockout-engineered heart tissue sensitive to metabolic stress such as serum withdrawal and restrictive feeding.CONCLUSION: The results suggest an important role of DNA methylation in the normal homeostasis of cardiomyocytes and during cardiac stress, which could make it an interesting target for cardiac therapy.

AB - BACKGROUND: DNA methylation acts as a mechanism of gene transcription regulation. It has recently gained attention as a possible therapeutic target in cardiac hypertrophy and heart failure. However, its exact role in cardiomyocytes remains controversial. Thus, we knocked out the main de novo DNA methyltransferase in cardiomyocytes, DNMT3A, in human induced pluripotent stem cells. Functional consequences of DNA methylation-deficiency under control and stress conditions were then assessed in human engineered heart tissue from knockout human induced pluripotent stem cell-derived cardiomyocytes.METHODS: DNMT3A was knocked out in human induced pluripotent stem cells by CRISPR/Cas9gene editing. Fibrin-based engineered heart tissue was generated from knockout and control human induced pluripotent stem cell-derived cardiomyocytes. Development and baseline contractility were analyzed by video-optical recording. Engineered heart tissue was subjected to different stress protocols, including serum starvation, serum variation, and restrictive feeding. Molecular, histological, and ultrastructural analyses were performed afterward.RESULTS: Knockout of DNMT3A in human cardiomyocytes had three main consequences for cardiomyocyte morphology and function: (1) Gene expression changes of contractile proteins such as higher atrial gene expression and lower MYH7/MYH6 ratio correlated with different contraction kinetics in knockout versus wild-type; (2) Aberrant activation of the glucose/lipid metabolism regulator peroxisome proliferator-activated receptor gamma was associated with accumulation of lipid vacuoles within knockout cardiomyocytes; (3) Hypoxia-inducible factor 1α protein instability was associated with impaired glucose metabolism and lower glycolytic enzyme expression, rendering knockout-engineered heart tissue sensitive to metabolic stress such as serum withdrawal and restrictive feeding.CONCLUSION: The results suggest an important role of DNA methylation in the normal homeostasis of cardiomyocytes and during cardiac stress, which could make it an interesting target for cardiac therapy.

U2 - 10.1161/CIRCULATIONAHA.119.044444

DO - 10.1161/CIRCULATIONAHA.119.044444

M3 - SCORING: Journal article

C2 - 32885664

VL - 142

SP - 1562

EP - 1578

JO - CIRCULATION

JF - CIRCULATION

SN - 0009-7322

IS - 16

ER -