Cargando…

PANoptosis is a prominent feature of desmoplakin cardiomyopathy

INTRODUCTION: Arrhythmogenic cardiomyopathy (ACM) is hereditary cardiomyopathy caused by pathogenic variants (mutations) in genes encoding the intercalated disc (ID), particularly desmosome proteins. ACM caused by mutations in the DSP gene encoding desmoplakin (DSP) is characterized by the prominenc...

Descripción completa

Detalles Bibliográficos
Autores principales: Olcum, Melis, Rouhi, Leila, Fan, Siyang, Gonzales, Maya M., Jeong, Hyun-Hwan, Zhao, Zhongming, Gurha, Priyatansh, Marian, Ali J.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9933912/
https://www.ncbi.nlm.nih.gov/pubmed/36818425
http://dx.doi.org/10.20517/jca.2022.34
_version_ 1784889769025601536
author Olcum, Melis
Rouhi, Leila
Fan, Siyang
Gonzales, Maya M.
Jeong, Hyun-Hwan
Zhao, Zhongming
Gurha, Priyatansh
Marian, Ali J.
author_facet Olcum, Melis
Rouhi, Leila
Fan, Siyang
Gonzales, Maya M.
Jeong, Hyun-Hwan
Zhao, Zhongming
Gurha, Priyatansh
Marian, Ali J.
author_sort Olcum, Melis
collection PubMed
description INTRODUCTION: Arrhythmogenic cardiomyopathy (ACM) is hereditary cardiomyopathy caused by pathogenic variants (mutations) in genes encoding the intercalated disc (ID), particularly desmosome proteins. ACM caused by mutations in the DSP gene encoding desmoplakin (DSP) is characterized by the prominence of cell death, myocardial fibrosis, and inflammation, and is referred to as desmoplakin cardiomyopathy. AIM: The aim of this article was to gain insight into the pathogenesis of DSP cardiomyopathy. METHODS AND RESULTS: The Dsp gene was exclusively deleted in cardiac myocytes using tamoxifen-inducible MerCreMer (Myh6-Mcm(Tam)) and floxed Dsp (Dsp(F/F)) mice (Myh6-Mcm(Tam):Dsp(F/F)). Recombination was induced upon subcutaneous injection of tamoxifen (30 mg/kg/d) for 5 days starting post-natal day 14. Survival was analyzed by Kaplan-Meier plots, cardiac function by echocardiography, arrhythmias by rhythm monitoring, and gene expression by RNA-Seq, immunoblotting, and immunofluorescence techniques. Cell death was analyzed by the TUNEL assay and the expression levels of specific markers were by RT-PCR and immunoblotting. Myocardial fibrosis was assessed by picrosirius red staining of the myocardial sections, RT-PCR, and immunoblotting. The Myh6-Mcm(Tam): Dsp(F/F) mice showed extensive molecular remodeling of the IDs and the differential expression of ~10,000 genes, which predicted activation of KDM5A, IRFs, and NFκB and suppression of PPARGC1A and RB1, among others in the DSP-deficient myocytes. Gene set enrichment analysis predicted activation of the TNFα/NFκB pathway, inflammation, cell death programs, and fibrosis. Analysis of cell death markers indicated PANoptosis, comprised of apoptosis (increased CASP3, CASP8, BAD and reduced BCL2), necroptosis (increased RIPK1, RIPK3, and MLKL), and pyroptosis (increased GSDMD and ASC or PYCARD) in the DSP-deficient myocytes. Transcript levels of the pro-inflammatory and pro-fibrotic genes were increased and myocardial fibrosis comprised ~25% of the myocardium in the DSP-deficient hearts. The Myh6-Mcm(Tam):Dsp(F/F) mice showed severe cardiac systolic dysfunction and ventricular arrhythmias, and died prematurely with a median survival rate of ~2 months. CONCLUSION: The findings identify PANoptosis as a prominent phenotypic feature of DSP cardiomyopathy and set the stage for delineating the specific molecular mechanisms involved in its pathogenesis. The model also provides the opportunity to test the effects of pharmacological and genetic interventions on myocardial fibrosis and cell death.
format Online
Article
Text
id pubmed-9933912
institution National Center for Biotechnology Information
language English
publishDate 2023
record_format MEDLINE/PubMed
spelling pubmed-99339122023-02-16 PANoptosis is a prominent feature of desmoplakin cardiomyopathy Olcum, Melis Rouhi, Leila Fan, Siyang Gonzales, Maya M. Jeong, Hyun-Hwan Zhao, Zhongming Gurha, Priyatansh Marian, Ali J. J Cardiovasc Aging Article INTRODUCTION: Arrhythmogenic cardiomyopathy (ACM) is hereditary cardiomyopathy caused by pathogenic variants (mutations) in genes encoding the intercalated disc (ID), particularly desmosome proteins. ACM caused by mutations in the DSP gene encoding desmoplakin (DSP) is characterized by the prominence of cell death, myocardial fibrosis, and inflammation, and is referred to as desmoplakin cardiomyopathy. AIM: The aim of this article was to gain insight into the pathogenesis of DSP cardiomyopathy. METHODS AND RESULTS: The Dsp gene was exclusively deleted in cardiac myocytes using tamoxifen-inducible MerCreMer (Myh6-Mcm(Tam)) and floxed Dsp (Dsp(F/F)) mice (Myh6-Mcm(Tam):Dsp(F/F)). Recombination was induced upon subcutaneous injection of tamoxifen (30 mg/kg/d) for 5 days starting post-natal day 14. Survival was analyzed by Kaplan-Meier plots, cardiac function by echocardiography, arrhythmias by rhythm monitoring, and gene expression by RNA-Seq, immunoblotting, and immunofluorescence techniques. Cell death was analyzed by the TUNEL assay and the expression levels of specific markers were by RT-PCR and immunoblotting. Myocardial fibrosis was assessed by picrosirius red staining of the myocardial sections, RT-PCR, and immunoblotting. The Myh6-Mcm(Tam): Dsp(F/F) mice showed extensive molecular remodeling of the IDs and the differential expression of ~10,000 genes, which predicted activation of KDM5A, IRFs, and NFκB and suppression of PPARGC1A and RB1, among others in the DSP-deficient myocytes. Gene set enrichment analysis predicted activation of the TNFα/NFκB pathway, inflammation, cell death programs, and fibrosis. Analysis of cell death markers indicated PANoptosis, comprised of apoptosis (increased CASP3, CASP8, BAD and reduced BCL2), necroptosis (increased RIPK1, RIPK3, and MLKL), and pyroptosis (increased GSDMD and ASC or PYCARD) in the DSP-deficient myocytes. Transcript levels of the pro-inflammatory and pro-fibrotic genes were increased and myocardial fibrosis comprised ~25% of the myocardium in the DSP-deficient hearts. The Myh6-Mcm(Tam):Dsp(F/F) mice showed severe cardiac systolic dysfunction and ventricular arrhythmias, and died prematurely with a median survival rate of ~2 months. CONCLUSION: The findings identify PANoptosis as a prominent phenotypic feature of DSP cardiomyopathy and set the stage for delineating the specific molecular mechanisms involved in its pathogenesis. The model also provides the opportunity to test the effects of pharmacological and genetic interventions on myocardial fibrosis and cell death. 2023-02 2023-01-01 /pmc/articles/PMC9933912/ /pubmed/36818425 http://dx.doi.org/10.20517/jca.2022.34 Text en https://creativecommons.org/licenses/by/4.0/Open Access This article is licensed under a Creative Commons Attribution 4. International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.
spellingShingle Article
Olcum, Melis
Rouhi, Leila
Fan, Siyang
Gonzales, Maya M.
Jeong, Hyun-Hwan
Zhao, Zhongming
Gurha, Priyatansh
Marian, Ali J.
PANoptosis is a prominent feature of desmoplakin cardiomyopathy
title PANoptosis is a prominent feature of desmoplakin cardiomyopathy
title_full PANoptosis is a prominent feature of desmoplakin cardiomyopathy
title_fullStr PANoptosis is a prominent feature of desmoplakin cardiomyopathy
title_full_unstemmed PANoptosis is a prominent feature of desmoplakin cardiomyopathy
title_short PANoptosis is a prominent feature of desmoplakin cardiomyopathy
title_sort panoptosis is a prominent feature of desmoplakin cardiomyopathy
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9933912/
https://www.ncbi.nlm.nih.gov/pubmed/36818425
http://dx.doi.org/10.20517/jca.2022.34
work_keys_str_mv AT olcummelis panoptosisisaprominentfeatureofdesmoplakincardiomyopathy
AT rouhileila panoptosisisaprominentfeatureofdesmoplakincardiomyopathy
AT fansiyang panoptosisisaprominentfeatureofdesmoplakincardiomyopathy
AT gonzalesmayam panoptosisisaprominentfeatureofdesmoplakincardiomyopathy
AT jeonghyunhwan panoptosisisaprominentfeatureofdesmoplakincardiomyopathy
AT zhaozhongming panoptosisisaprominentfeatureofdesmoplakincardiomyopathy
AT gurhapriyatansh panoptosisisaprominentfeatureofdesmoplakincardiomyopathy
AT marianalij panoptosisisaprominentfeatureofdesmoplakincardiomyopathy