Cargando…

Distinct transcriptomic changes in E14.5 mouse skeletal muscle lacking RYR1 or Ca(v)1.1 converge at E18.5

In skeletal muscle the coordinated actions of two mechanically coupled Ca(2+) channels—the 1,4-dihydropyridine receptor (Ca(v)1.1) and the type 1 ryanodine receptor (RYR1)–underlie the molecular mechanism of rapid cytosolic [Ca(2+)] increase leading to contraction. While both [Ca(2+)](i) and contrac...

Descripción completa

Detalles Bibliográficos
Autores principales: Filipova, Dilyana, Henry, Margit, Rotshteyn, Tamara, Brunn, Anna, Carstov, Mariana, Deckert, Martina, Hescheler, Jürgen, Sachinidis, Agapios, Pfitzer, Gabriele, Papadopoulos, Symeon
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5854361/
https://www.ncbi.nlm.nih.gov/pubmed/29543863
http://dx.doi.org/10.1371/journal.pone.0194428
_version_ 1783306901418672128
author Filipova, Dilyana
Henry, Margit
Rotshteyn, Tamara
Brunn, Anna
Carstov, Mariana
Deckert, Martina
Hescheler, Jürgen
Sachinidis, Agapios
Pfitzer, Gabriele
Papadopoulos, Symeon
author_facet Filipova, Dilyana
Henry, Margit
Rotshteyn, Tamara
Brunn, Anna
Carstov, Mariana
Deckert, Martina
Hescheler, Jürgen
Sachinidis, Agapios
Pfitzer, Gabriele
Papadopoulos, Symeon
author_sort Filipova, Dilyana
collection PubMed
description In skeletal muscle the coordinated actions of two mechanically coupled Ca(2+) channels—the 1,4-dihydropyridine receptor (Ca(v)1.1) and the type 1 ryanodine receptor (RYR1)–underlie the molecular mechanism of rapid cytosolic [Ca(2+)] increase leading to contraction. While both [Ca(2+)](i) and contractile activity have been implicated in the regulation of myogenesis, less is known about potential specific roles of Ca(v)1.1 and RYR1 in skeletal muscle development. In this study, we analyzed the histology and the transcriptomic changes occurring at E14.5 –the end of primary myogenesis and around the onset of intrauterine limb movement, and at E18.5 –the end of secondary myogenesis, in WT, RYR1(-/-), and Ca(v)1.1(-/-) murine limb skeletal muscle. At E14.5 the muscle histology of both mutants exhibited initial alterations, which became much more severe at E18.5. Immunohistological analysis also revealed higher levels of activated caspase-3 in the Ca(v)1.1(-/-) muscles at E14.5, indicating an increase in apoptosis. With WT littermates as controls, microarray analyses identified 61 and 97 differentially regulated genes (DEGs) at E14.5, and 493 and 1047 DEGs at E18.5, in RYR1(-/-) and Ca(v)1.1(-/-) samples, respectively. Gene enrichment analysis detected no overlap in the affected biological processes and pathways in the two mutants at E14.5, whereas at E18.5 there was a significant overlap of DEGs in both mutants, affecting predominantly processes linked to muscle contraction. Moreover, the E18.5 vs. E14.5 comparison revealed multiple genotype-specific DEGs involved in contraction, cell cycle and miRNA-mediated signaling in WT, neuronal and bone development in RYR1(-/-), and lipid metabolism in Ca(v)1.1(-/-) samples. Taken together, our study reveals discrete changes in the global transcriptome occurring in limb skeletal muscle from E14.5 to E18.5 in WT, RYR1(-/-) and Ca(v)1.1(-/-) mice. Our results suggest distinct functional roles for RYR1 and Ca(v)1.1 in skeletal primary and secondary myogenesis.
format Online
Article
Text
id pubmed-5854361
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-58543612018-03-28 Distinct transcriptomic changes in E14.5 mouse skeletal muscle lacking RYR1 or Ca(v)1.1 converge at E18.5 Filipova, Dilyana Henry, Margit Rotshteyn, Tamara Brunn, Anna Carstov, Mariana Deckert, Martina Hescheler, Jürgen Sachinidis, Agapios Pfitzer, Gabriele Papadopoulos, Symeon PLoS One Research Article In skeletal muscle the coordinated actions of two mechanically coupled Ca(2+) channels—the 1,4-dihydropyridine receptor (Ca(v)1.1) and the type 1 ryanodine receptor (RYR1)–underlie the molecular mechanism of rapid cytosolic [Ca(2+)] increase leading to contraction. While both [Ca(2+)](i) and contractile activity have been implicated in the regulation of myogenesis, less is known about potential specific roles of Ca(v)1.1 and RYR1 in skeletal muscle development. In this study, we analyzed the histology and the transcriptomic changes occurring at E14.5 –the end of primary myogenesis and around the onset of intrauterine limb movement, and at E18.5 –the end of secondary myogenesis, in WT, RYR1(-/-), and Ca(v)1.1(-/-) murine limb skeletal muscle. At E14.5 the muscle histology of both mutants exhibited initial alterations, which became much more severe at E18.5. Immunohistological analysis also revealed higher levels of activated caspase-3 in the Ca(v)1.1(-/-) muscles at E14.5, indicating an increase in apoptosis. With WT littermates as controls, microarray analyses identified 61 and 97 differentially regulated genes (DEGs) at E14.5, and 493 and 1047 DEGs at E18.5, in RYR1(-/-) and Ca(v)1.1(-/-) samples, respectively. Gene enrichment analysis detected no overlap in the affected biological processes and pathways in the two mutants at E14.5, whereas at E18.5 there was a significant overlap of DEGs in both mutants, affecting predominantly processes linked to muscle contraction. Moreover, the E18.5 vs. E14.5 comparison revealed multiple genotype-specific DEGs involved in contraction, cell cycle and miRNA-mediated signaling in WT, neuronal and bone development in RYR1(-/-), and lipid metabolism in Ca(v)1.1(-/-) samples. Taken together, our study reveals discrete changes in the global transcriptome occurring in limb skeletal muscle from E14.5 to E18.5 in WT, RYR1(-/-) and Ca(v)1.1(-/-) mice. Our results suggest distinct functional roles for RYR1 and Ca(v)1.1 in skeletal primary and secondary myogenesis. Public Library of Science 2018-03-15 /pmc/articles/PMC5854361/ /pubmed/29543863 http://dx.doi.org/10.1371/journal.pone.0194428 Text en © 2018 Filipova et al http://creativecommons.org/licenses/by/4.0/ This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/) , which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Article
Filipova, Dilyana
Henry, Margit
Rotshteyn, Tamara
Brunn, Anna
Carstov, Mariana
Deckert, Martina
Hescheler, Jürgen
Sachinidis, Agapios
Pfitzer, Gabriele
Papadopoulos, Symeon
Distinct transcriptomic changes in E14.5 mouse skeletal muscle lacking RYR1 or Ca(v)1.1 converge at E18.5
title Distinct transcriptomic changes in E14.5 mouse skeletal muscle lacking RYR1 or Ca(v)1.1 converge at E18.5
title_full Distinct transcriptomic changes in E14.5 mouse skeletal muscle lacking RYR1 or Ca(v)1.1 converge at E18.5
title_fullStr Distinct transcriptomic changes in E14.5 mouse skeletal muscle lacking RYR1 or Ca(v)1.1 converge at E18.5
title_full_unstemmed Distinct transcriptomic changes in E14.5 mouse skeletal muscle lacking RYR1 or Ca(v)1.1 converge at E18.5
title_short Distinct transcriptomic changes in E14.5 mouse skeletal muscle lacking RYR1 or Ca(v)1.1 converge at E18.5
title_sort distinct transcriptomic changes in e14.5 mouse skeletal muscle lacking ryr1 or ca(v)1.1 converge at e18.5
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5854361/
https://www.ncbi.nlm.nih.gov/pubmed/29543863
http://dx.doi.org/10.1371/journal.pone.0194428
work_keys_str_mv AT filipovadilyana distincttranscriptomicchangesine145mouseskeletalmusclelackingryr1orcav11convergeate185
AT henrymargit distincttranscriptomicchangesine145mouseskeletalmusclelackingryr1orcav11convergeate185
AT rotshteyntamara distincttranscriptomicchangesine145mouseskeletalmusclelackingryr1orcav11convergeate185
AT brunnanna distincttranscriptomicchangesine145mouseskeletalmusclelackingryr1orcav11convergeate185
AT carstovmariana distincttranscriptomicchangesine145mouseskeletalmusclelackingryr1orcav11convergeate185
AT deckertmartina distincttranscriptomicchangesine145mouseskeletalmusclelackingryr1orcav11convergeate185
AT heschelerjurgen distincttranscriptomicchangesine145mouseskeletalmusclelackingryr1orcav11convergeate185
AT sachinidisagapios distincttranscriptomicchangesine145mouseskeletalmusclelackingryr1orcav11convergeate185
AT pfitzergabriele distincttranscriptomicchangesine145mouseskeletalmusclelackingryr1orcav11convergeate185
AT papadopoulossymeon distincttranscriptomicchangesine145mouseskeletalmusclelackingryr1orcav11convergeate185