Cargando…

Indirect epigenetic testing identifies a diagnostic signature of cardiomyocyte DNA methylation in heart failure

Precision-based molecular phenotyping of heart failure must overcome limited access to cardiac tissue. Although epigenetic alterations have been found to underlie pathological cardiac gene dysregulation, the clinical utility of myocardial epigenomics remains narrow owing to limited clinical access t...

Descripción completa

Detalles Bibliográficos
Autores principales: Oeing, Christian U., Pepin, Mark E., Saul, Kerstin B., Agircan, Ayça Seyhan, Assenov, Yassen, Merkel, Tobias S., Sedaghat-Hamedani, Farbod, Weis, Tanja, Meder, Benjamin, Guan, Kaomei, Plass, Christoph, Weichenhan, Dieter, Siede, Dominik, Backs, Johannes
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Springer Berlin Heidelberg 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10027651/
https://www.ncbi.nlm.nih.gov/pubmed/36939901
http://dx.doi.org/10.1007/s00395-022-00954-3
_version_ 1784909747507429376
author Oeing, Christian U.
Pepin, Mark E.
Saul, Kerstin B.
Agircan, Ayça Seyhan
Assenov, Yassen
Merkel, Tobias S.
Sedaghat-Hamedani, Farbod
Weis, Tanja
Meder, Benjamin
Guan, Kaomei
Plass, Christoph
Weichenhan, Dieter
Siede, Dominik
Backs, Johannes
author_facet Oeing, Christian U.
Pepin, Mark E.
Saul, Kerstin B.
Agircan, Ayça Seyhan
Assenov, Yassen
Merkel, Tobias S.
Sedaghat-Hamedani, Farbod
Weis, Tanja
Meder, Benjamin
Guan, Kaomei
Plass, Christoph
Weichenhan, Dieter
Siede, Dominik
Backs, Johannes
author_sort Oeing, Christian U.
collection PubMed
description Precision-based molecular phenotyping of heart failure must overcome limited access to cardiac tissue. Although epigenetic alterations have been found to underlie pathological cardiac gene dysregulation, the clinical utility of myocardial epigenomics remains narrow owing to limited clinical access to tissue. Therefore, the current study determined whether patient plasma confers indirect phenotypic, transcriptional, and/or epigenetic alterations to ex vivo cardiomyocytes to mirror the failing human myocardium. Neonatal rat ventricular myocytes (NRVMs) and single-origin human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and were treated with blood plasma samples from patients with dilated cardiomyopathy (DCM) and donor subjects lacking history of cardiovascular disease. Following plasma treatments, NRVMs and hiPSC-CMs underwent significant hypertrophy relative to non-failing controls, as determined via automated high-content screening. Array-based DNA methylation analysis of plasma-treated hiPSC-CMs and cardiac biopsies uncovered robust, and conserved, alterations in cardiac DNA methylation, from which 100 sites were validated using an independent cohort. Among the CpG sites identified, hypo-methylation of the ATG promoter was identified as a diagnostic marker of HF, wherein cg03800765 methylation (AUC = 0.986, P < 0.0001) was found to out-perform circulating NT-proBNP levels in differentiating heart failure. Taken together, these findings support a novel approach of indirect epigenetic testing in human HF. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s00395-022-00954-3.
format Online
Article
Text
id pubmed-10027651
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Springer Berlin Heidelberg
record_format MEDLINE/PubMed
spelling pubmed-100276512023-03-22 Indirect epigenetic testing identifies a diagnostic signature of cardiomyocyte DNA methylation in heart failure Oeing, Christian U. Pepin, Mark E. Saul, Kerstin B. Agircan, Ayça Seyhan Assenov, Yassen Merkel, Tobias S. Sedaghat-Hamedani, Farbod Weis, Tanja Meder, Benjamin Guan, Kaomei Plass, Christoph Weichenhan, Dieter Siede, Dominik Backs, Johannes Basic Res Cardiol Original Contribution Precision-based molecular phenotyping of heart failure must overcome limited access to cardiac tissue. Although epigenetic alterations have been found to underlie pathological cardiac gene dysregulation, the clinical utility of myocardial epigenomics remains narrow owing to limited clinical access to tissue. Therefore, the current study determined whether patient plasma confers indirect phenotypic, transcriptional, and/or epigenetic alterations to ex vivo cardiomyocytes to mirror the failing human myocardium. Neonatal rat ventricular myocytes (NRVMs) and single-origin human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and were treated with blood plasma samples from patients with dilated cardiomyopathy (DCM) and donor subjects lacking history of cardiovascular disease. Following plasma treatments, NRVMs and hiPSC-CMs underwent significant hypertrophy relative to non-failing controls, as determined via automated high-content screening. Array-based DNA methylation analysis of plasma-treated hiPSC-CMs and cardiac biopsies uncovered robust, and conserved, alterations in cardiac DNA methylation, from which 100 sites were validated using an independent cohort. Among the CpG sites identified, hypo-methylation of the ATG promoter was identified as a diagnostic marker of HF, wherein cg03800765 methylation (AUC = 0.986, P < 0.0001) was found to out-perform circulating NT-proBNP levels in differentiating heart failure. Taken together, these findings support a novel approach of indirect epigenetic testing in human HF. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s00395-022-00954-3. Springer Berlin Heidelberg 2023-03-20 2023 /pmc/articles/PMC10027651/ /pubmed/36939901 http://dx.doi.org/10.1007/s00395-022-00954-3 Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Original Contribution
Oeing, Christian U.
Pepin, Mark E.
Saul, Kerstin B.
Agircan, Ayça Seyhan
Assenov, Yassen
Merkel, Tobias S.
Sedaghat-Hamedani, Farbod
Weis, Tanja
Meder, Benjamin
Guan, Kaomei
Plass, Christoph
Weichenhan, Dieter
Siede, Dominik
Backs, Johannes
Indirect epigenetic testing identifies a diagnostic signature of cardiomyocyte DNA methylation in heart failure
title Indirect epigenetic testing identifies a diagnostic signature of cardiomyocyte DNA methylation in heart failure
title_full Indirect epigenetic testing identifies a diagnostic signature of cardiomyocyte DNA methylation in heart failure
title_fullStr Indirect epigenetic testing identifies a diagnostic signature of cardiomyocyte DNA methylation in heart failure
title_full_unstemmed Indirect epigenetic testing identifies a diagnostic signature of cardiomyocyte DNA methylation in heart failure
title_short Indirect epigenetic testing identifies a diagnostic signature of cardiomyocyte DNA methylation in heart failure
title_sort indirect epigenetic testing identifies a diagnostic signature of cardiomyocyte dna methylation in heart failure
topic Original Contribution
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10027651/
https://www.ncbi.nlm.nih.gov/pubmed/36939901
http://dx.doi.org/10.1007/s00395-022-00954-3
work_keys_str_mv AT oeingchristianu indirectepigenetictestingidentifiesadiagnosticsignatureofcardiomyocytednamethylationinheartfailure
AT pepinmarke indirectepigenetictestingidentifiesadiagnosticsignatureofcardiomyocytednamethylationinheartfailure
AT saulkerstinb indirectepigenetictestingidentifiesadiagnosticsignatureofcardiomyocytednamethylationinheartfailure
AT agircanaycaseyhan indirectepigenetictestingidentifiesadiagnosticsignatureofcardiomyocytednamethylationinheartfailure
AT assenovyassen indirectepigenetictestingidentifiesadiagnosticsignatureofcardiomyocytednamethylationinheartfailure
AT merkeltobiass indirectepigenetictestingidentifiesadiagnosticsignatureofcardiomyocytednamethylationinheartfailure
AT sedaghathamedanifarbod indirectepigenetictestingidentifiesadiagnosticsignatureofcardiomyocytednamethylationinheartfailure
AT weistanja indirectepigenetictestingidentifiesadiagnosticsignatureofcardiomyocytednamethylationinheartfailure
AT mederbenjamin indirectepigenetictestingidentifiesadiagnosticsignatureofcardiomyocytednamethylationinheartfailure
AT guankaomei indirectepigenetictestingidentifiesadiagnosticsignatureofcardiomyocytednamethylationinheartfailure
AT plasschristoph indirectepigenetictestingidentifiesadiagnosticsignatureofcardiomyocytednamethylationinheartfailure
AT weichenhandieter indirectepigenetictestingidentifiesadiagnosticsignatureofcardiomyocytednamethylationinheartfailure
AT siededominik indirectepigenetictestingidentifiesadiagnosticsignatureofcardiomyocytednamethylationinheartfailure
AT backsjohannes indirectepigenetictestingidentifiesadiagnosticsignatureofcardiomyocytednamethylationinheartfailure