Transcriptome signature of ventricular arrhythmia in dilated cardiomyopathy reveals increased fibrosis and activated TP53

https://doi.org/10.1016/j.yjmcc.2019.12.010Get rights and content

Highlights

  • About 1:3 of dilated cardiomyopathy patients experience ventricular tachycardia.

  • Our understanding of the biological basis of these arrhythmias is incomplete.

  • RNA-seq demonstrates discrete arrhythmia and non-arrhythmia expression profiles.

  • These expression signatures are enriched for distinct biological pathways.

  • TP53 and TGFβ1 pathways distinguish arrhythmia and non-arrhythmia cohorts.

Abstract

Aims

One-third of DCM patients experience ventricular tachycardia (VT), but a clear biological basis for this has not been established. The purpose of this study was to identify transcriptome signatures and enriched pathways in the hearts of dilated cardiomyopathy (DCM) patients with VT.

Methods and results

We used RNA-sequencing in explanted heart tissue from 49 samples: 19 DCM patients with VT, 16 DCM patients without VT, and 14 non-failing controls. We compared each DCM cohort to the controls and identified the genes that were differentially expressed in DCM patients with VT but not without VT. Differentially expressed genes were evaluated using pathway analysis, and pathways of interest were investigated by qRT-PCR validation, Western blot, and microscopy. There were 590 genes differentially expressed in DCM patients with VT that are not differentially expressed in patients without VT. These genes were enriched for genes in the TGFß1 and TP53 signaling pathways. Increased fibrosis and activated TP53 signaling was demonstrated in heart tissue of DCM patients with VT.

Conclusions

Our study supports that distinct biological mechanisms distinguish ventricular arrhythmia in DCM patients.

Introduction

Cardiac arrhythmias frequently complicate dilated cardiomyopathy (DCM), causing physical symptoms, a need for medications and/or device therapy, and sudden cardiac death [1]. In DCM, life-threatening arrhythmias can present early in disease [2], and approximately one-third of patients experience frequent ventricular arrhythmias, which may occur independent of left ventricular (LV) dysfunction [3]. Although arrhythmia-prone patients can be identified and stratified clinically once arrhythmias develop, there is a significant knowledge gap in understanding the biological mechanisms that contribute to ventricular arrhythmias in arrhythmogenic DCM patients.

Molecular and histological studies of human heart tissue from DCM patients with and without history of ventricular arrhythmias showed that patients with a positive history had increased hypertrophy, evidence of oxidative stress, and fibrosis. Microarray analysis showed differences in gene expression that included increased expression of select genes encoding extracellular matrix proteins (FGF18, COL4A2, COL12A1) and ion channels (KCNN2, TRPM7); a formal analysis for enrichment of differentially expressed genes in key biological pathways has not been performed and prior results were not compared against non-failing (NF) controls [4].

We and others have previously showed that although there is a common heart failure gene expression signature for heart failure, there are also distinct gene expression signatures that distinguish DCM from ischemic cardiomyopathy that suggest distinct disease mechanisms [[5], [6], [7], [8], [9]]. Here, we hypothesized that within DCM, sub-phenotypic gene expression signatures exist that distinguish the arrhythmogenic DCM (aDCM) from the non-arrhythmogenic DCM (naDCM) state. To test this hypothesis, we performed RNA-sequencing (RNA-seq) on 49 explanted human hearts and used a multi-analytic approach to discern aDCM- and naDCM-specific expression profiles. Using pathway analysis, our results support that distinct disease mechanisms exist within DCM that separate arrhythmogenic and non-arrhythmogenic biological sub-phenotypes of DCM, including activation of TGFβ1 and TP53 signaling.

Section snippets

Tissue collection

Heart tissues were collected as described previously [9]. Briefly, the Colorado Multicenter Institutional Review Board approved the protocol for the collection, storage, and analysis of human tissue at the University of Colorado Hospital as part of the Division of Cardiology Cardiac Tissue Biobank (COMIRB, protocol 01-568). Transplant-listed patients signed written consent for use of their explanted hearts for research purposes. Explanted tissue, absent of scarring or infarcted segments, was

Clinical characteristics of patients

Forty-nine human hearts were investigated from: 19 aDCM patients, 16 naDCM patients, and 14 NF (Additional File 1). Clinical criteria for aDCM and naDCM patients are described in the Methods. Table 1 summarizes clinical characteristics between the patient groups. As expected, the aDCM cohort had a significantly greater proportion of patients taking amiodarone (p = .04), having an implantable cardioverter defibrillator (ICD; p = .01), and experiencing ventricular tachycardia (p < .00001).

Principal component analysis of the cohorts

To

aDCM and naDCM have distinct gene expression signatures

Prior studies have identified gene expression changes that occur in the failing human heart and are distinct from the healthy state, and that failing hearts share common gene expression signatures. However, differences in gene expression exist within heart failure that are able to accurately classify hearts by underlying disease etiologies, such as ischemic or non-ischemic disease [9]. Here, we showed that once the common, overlapping expression shared by all DCM patients is removed, we can

Conclusions

We used RNA-seq, pathway analysis, histology, and molecular studies to demonstrate that clinical distinctions in DCM arrhythmogenic sub-phenotypes result in biologically relevant differences. In our study, aDCM and naDCM were revealed to have distinct gene expression signatures, including significant differences in the TGFβ1 and TP53 pathways, which were validated by dysregulated protein expression and histological evidence of differing degrees of fibrosis. Notably, explanted LV tissue

Funding

This work was supported by the National Institutes of Health [UL1 TR002535, R01 HL69071, R01 116906 (LM), R01 HL109209 (MRGT)] and the Fondation Leducq [14-CVD 03]. Contents are the authors' sole responsibility and do not necessarily represent official NIH views.

Declaration of competing interest

None.

Acknowledgements

The authors would like to thank Dr. Peter Buttrick and the University of Colorado's Division of Cardiology for ongoing maintenance of the human cardiac tissue biobank as well as the patients and organ donors who provided the heart tissues used in this study.

References (67)

  • W.W. Brooks et al.

    Myocardial fibrosis in transforming growth factor beta(1)heterozygous mice

    J. Mol. Cell. Cardiol.

    (2000)
  • K.M. Hyman

    Transforming growth factor-beta1 induces apoptosis in vascular endothelial cells by activation of mitogen-activated protein kinase

    Surgery

    (2002)
  • Y. Yoshida

    p53-Induced inflammation exacerbates cardiac dysfunction during pressure overload

    J. Mol. Cell. Cardiol.

    (2015)
  • H. Song

    Increased p53 protein expression in human failing myocardium

    J. Heart Lung Transplant.

    (1999)
  • O. Bergmann

    Dynamics of cell generation and turnover in the human heart

    Cell

    (2015)
  • C.B. Hensley

    Amiodarone decreases Na,K-ATPase alpha 2 and beta 2 expression specifically in cardiac ventricle

    J. Mol. Cell. Cardiol.

    (1994)
  • E.C. Stecker

    Public health burden of sudden cardiac death in the United States

    Circ. Arrhythm. Electrophysiol.

    (2014)
  • A. Spezzacatene

    Arrhythmogenic phenotype in dilated cardiomyopathy: natural history and predictors of life-threatening arrhythmias

    J. Am. Heart Assoc.

    (2015)
  • N. Parajuli

    Determinants of ventricular arrhythmias in human explanted hearts with dilated cardiomyopathy

    Eur. J. Clin. Investig.

    (2015)
  • M.M. Kittleson

    Gene expression analysis of ischemic and nonischemic cardiomyopathy: shared and distinct genes in the development of heart failure

    Physiol. Genomics

    (2005)
  • R. Kuner

    Genomic analysis reveals poor separation of human cardiomyopathies of ischemic and nonischemic etiologies

    Physiol. Genomics

    (2008)
  • M.M. Kittleson

    Identification of a gene expression profile that differentiates between ischemic and nonischemic cardiomyopathy

    Circulation

    (2004)
  • M.E. Sweet

    Transcriptome analysis of human heart failure reveals dysregulated cell adhesion in dilated cardiomyopathy and activated immune pathways in ischemic heart failure

    BMC Genomics

    (2018)
  • T.D. Wu et al.

    Fast and SNP-tolerant detection of complex variants and splicing in short reads

    Bioinformatics

    (2010)
  • C. Trapnell

    Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation

    Nat. Biotechnol.

    (2010)
  • K.C. Yang

    Deep RNA sequencing reveals dynamic regulation of myocardial noncoding RNAs in failing human heart and remodeling with mechanical circulatory support

    Circulation

    (2014)
  • D. Szklarczyk

    STRING v10: protein-protein interaction networks, integrated over the tree of life

    Nucleic Acids Res.

    (2015)
  • S. Roy

    miR-30c and miR-193 are a part of the TGF-beta-dependent regulatory network controlling extracellular matrix genes in liver fibrosis

    J. Dig. Dis.

    (2015)
  • S. Carbon

    AmiGO: online access to ontology and annotation data

    Bioinformatics

    (2009)
  • H. Mi

    PANTHER version 14: more genomes, a new PANTHER GO-slim and improvements in enrichment analysis tools

    Nucleic Acids Res.

    (2019)
  • C. The Gene Ontology

    The gene ontology resource: 20 years and still GOing strong

    Nucleic Acids Res.

    (2019)
  • S.A. Doppler

    Cardiac fibroblasts: more than mechanical support

    J. Thorac. Dis.

    (2017)
  • J.E. Friedman

    Pyrroloquinoline quinone prevents developmental programming of microbial dysbiosis and macrophage polarization to attenuate liver fibrosis in offspring of obese mice

    Hepatol. Commun.

    (2018)
  • Cited by (18)

    • The Human Explanted Heart Program: A translational bridge for cardiovascular medicine

      2021, Biochimica et Biophysica Acta - Molecular Basis of Disease
      Citation Excerpt :

      In addition, RNA-Seq analysis provides an improved and expanded RNA profiling with higher specificity and sensitivity, especially for less abundant transcripts. RNA expression profiling has provided new insight into disease pathogenesis including transcriptome signatures of DCM hearts with ventricular arrhythmia [36,92–95]; however, full knowledge of the broader repertoire of healthy hearts' molecular and genetic landscape is fundamental. To achieve that, we profiled both cells and nuclei from six distinct cardiac anatomical regions (apex, septum, left/right atria and ventricles) of the non-failing donor hearts, and constructed a most comprehensive transcriptome atlas as the reference framework to advance mechanistic exploration into heart diseases (Fig. 5E) [36,37,93–96].

    • Mechanism of Xuefu Zhuyu Tang in Myocardial Infarction Treatment Based on Network Pharmacology

      2023, Proceedings - 2023 2023 IEEE International Conference on Bioinformatics and Biomedicine, BIBM 2023
    View all citing articles on Scopus
    View full text