Cargando…

Transcriptional Profiling of Cultured, Embryonic Epicardial Cells Identifies Novel Genes and Signaling Pathways Regulated by TGFβR3 In Vitro

The epicardium plays an important role in coronary vessel formation and Tgfbr3(-/-) mice exhibit failed coronary vessel development associated with decreased epicardial cell invasion. Immortalized Tgfbr3(-/-) epicardial cells display the same defects. Tgfbr3(+/+) and Tgfbr3(-/-) cells incubated for...

Descripción completa

Detalles Bibliográficos
Autores principales: DeLaughter, Daniel M., Clark, Cynthia R., Christodoulou, Danos C., Seidman, Christine E., Baldwin, H. Scott, Seidman, J. G., Barnett, Joey V.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4978490/
https://www.ncbi.nlm.nih.gov/pubmed/27505173
http://dx.doi.org/10.1371/journal.pone.0159710
_version_ 1782447182756970496
author DeLaughter, Daniel M.
Clark, Cynthia R.
Christodoulou, Danos C.
Seidman, Christine E.
Baldwin, H. Scott
Seidman, J. G.
Barnett, Joey V.
author_facet DeLaughter, Daniel M.
Clark, Cynthia R.
Christodoulou, Danos C.
Seidman, Christine E.
Baldwin, H. Scott
Seidman, J. G.
Barnett, Joey V.
author_sort DeLaughter, Daniel M.
collection PubMed
description The epicardium plays an important role in coronary vessel formation and Tgfbr3(-/-) mice exhibit failed coronary vessel development associated with decreased epicardial cell invasion. Immortalized Tgfbr3(-/-) epicardial cells display the same defects. Tgfbr3(+/+) and Tgfbr3(-/-) cells incubated for 72 hours with VEH or ligands known to promote invasion via TGFβR3 (TGFβ1, TGFβ2, BMP2), for 72 hours were harvested for RNA-seq analysis. We selected for genes >2-fold differentially expressed between Tgfbr3(+/+) and Tgfbr3(-/-) cells when incubated with VEH (604), TGFβ1 (515), TGFβ2 (553), or BMP2 (632). Gene Ontology (GO) analysis of these genes identified dysregulated biological processes consistent with the defects observed in Tgfbr3(-/-) cells, including those associated with extracellular matrix interaction. GO and Gene Regulatory Network (GRN) analysis identified distinct expression profiles between TGFβ1-TGFβ2 and VEH-BMP2 incubated cells, consistent with the differential response of epicardial cells to these ligands in vitro. Despite the differences observed between Tgfbr3(+/+) and Tgfbr3(-/-) cells after TGFβ and BMP ligand addition, GRNs constructed from these gene lists identified NF-ĸB as a key nodal point for all ligands examined. Tgfbr3(-/-) cells exhibited decreased expression of genes known to be activated by NF-ĸB signaling. NF-ĸB activity was stimulated in Tgfbr3(+/+) epicardial cells after TGFβ2 or BMP2 incubation, while Tgfbr3(-/-) cells failed to activate NF-ĸB in response to these ligands. Tgfbr3(+/+) epicardial cells incubated with an inhibitor of NF-ĸB signaling no longer invaded into a collagen gel in response to TGFβ2 or BMP2. These data suggest that NF-ĸB signaling is dysregulated in Tgfbr3(-/-) epicardial cells and that NF-ĸB signaling is required for epicardial cell invasion in vitro. Our approach successfully identified a signaling pathway important in epicardial cell behavior downstream of TGFβR3. Overall, the genes and signaling pathways identified through our analysis yield the first comprehensive list of candidate genes whose expression is dependent on TGFβR3 signaling.
format Online
Article
Text
id pubmed-4978490
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-49784902016-08-25 Transcriptional Profiling of Cultured, Embryonic Epicardial Cells Identifies Novel Genes and Signaling Pathways Regulated by TGFβR3 In Vitro DeLaughter, Daniel M. Clark, Cynthia R. Christodoulou, Danos C. Seidman, Christine E. Baldwin, H. Scott Seidman, J. G. Barnett, Joey V. PLoS One Research Article The epicardium plays an important role in coronary vessel formation and Tgfbr3(-/-) mice exhibit failed coronary vessel development associated with decreased epicardial cell invasion. Immortalized Tgfbr3(-/-) epicardial cells display the same defects. Tgfbr3(+/+) and Tgfbr3(-/-) cells incubated for 72 hours with VEH or ligands known to promote invasion via TGFβR3 (TGFβ1, TGFβ2, BMP2), for 72 hours were harvested for RNA-seq analysis. We selected for genes >2-fold differentially expressed between Tgfbr3(+/+) and Tgfbr3(-/-) cells when incubated with VEH (604), TGFβ1 (515), TGFβ2 (553), or BMP2 (632). Gene Ontology (GO) analysis of these genes identified dysregulated biological processes consistent with the defects observed in Tgfbr3(-/-) cells, including those associated with extracellular matrix interaction. GO and Gene Regulatory Network (GRN) analysis identified distinct expression profiles between TGFβ1-TGFβ2 and VEH-BMP2 incubated cells, consistent with the differential response of epicardial cells to these ligands in vitro. Despite the differences observed between Tgfbr3(+/+) and Tgfbr3(-/-) cells after TGFβ and BMP ligand addition, GRNs constructed from these gene lists identified NF-ĸB as a key nodal point for all ligands examined. Tgfbr3(-/-) cells exhibited decreased expression of genes known to be activated by NF-ĸB signaling. NF-ĸB activity was stimulated in Tgfbr3(+/+) epicardial cells after TGFβ2 or BMP2 incubation, while Tgfbr3(-/-) cells failed to activate NF-ĸB in response to these ligands. Tgfbr3(+/+) epicardial cells incubated with an inhibitor of NF-ĸB signaling no longer invaded into a collagen gel in response to TGFβ2 or BMP2. These data suggest that NF-ĸB signaling is dysregulated in Tgfbr3(-/-) epicardial cells and that NF-ĸB signaling is required for epicardial cell invasion in vitro. Our approach successfully identified a signaling pathway important in epicardial cell behavior downstream of TGFβR3. Overall, the genes and signaling pathways identified through our analysis yield the first comprehensive list of candidate genes whose expression is dependent on TGFβR3 signaling. Public Library of Science 2016-08-09 /pmc/articles/PMC4978490/ /pubmed/27505173 http://dx.doi.org/10.1371/journal.pone.0159710 Text en © 2016 DeLaughter 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
DeLaughter, Daniel M.
Clark, Cynthia R.
Christodoulou, Danos C.
Seidman, Christine E.
Baldwin, H. Scott
Seidman, J. G.
Barnett, Joey V.
Transcriptional Profiling of Cultured, Embryonic Epicardial Cells Identifies Novel Genes and Signaling Pathways Regulated by TGFβR3 In Vitro
title Transcriptional Profiling of Cultured, Embryonic Epicardial Cells Identifies Novel Genes and Signaling Pathways Regulated by TGFβR3 In Vitro
title_full Transcriptional Profiling of Cultured, Embryonic Epicardial Cells Identifies Novel Genes and Signaling Pathways Regulated by TGFβR3 In Vitro
title_fullStr Transcriptional Profiling of Cultured, Embryonic Epicardial Cells Identifies Novel Genes and Signaling Pathways Regulated by TGFβR3 In Vitro
title_full_unstemmed Transcriptional Profiling of Cultured, Embryonic Epicardial Cells Identifies Novel Genes and Signaling Pathways Regulated by TGFβR3 In Vitro
title_short Transcriptional Profiling of Cultured, Embryonic Epicardial Cells Identifies Novel Genes and Signaling Pathways Regulated by TGFβR3 In Vitro
title_sort transcriptional profiling of cultured, embryonic epicardial cells identifies novel genes and signaling pathways regulated by tgfβr3 in vitro
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4978490/
https://www.ncbi.nlm.nih.gov/pubmed/27505173
http://dx.doi.org/10.1371/journal.pone.0159710
work_keys_str_mv AT delaughterdanielm transcriptionalprofilingofculturedembryonicepicardialcellsidentifiesnovelgenesandsignalingpathwaysregulatedbytgfbr3invitro
AT clarkcynthiar transcriptionalprofilingofculturedembryonicepicardialcellsidentifiesnovelgenesandsignalingpathwaysregulatedbytgfbr3invitro
AT christodouloudanosc transcriptionalprofilingofculturedembryonicepicardialcellsidentifiesnovelgenesandsignalingpathwaysregulatedbytgfbr3invitro
AT seidmanchristinee transcriptionalprofilingofculturedembryonicepicardialcellsidentifiesnovelgenesandsignalingpathwaysregulatedbytgfbr3invitro
AT baldwinhscott transcriptionalprofilingofculturedembryonicepicardialcellsidentifiesnovelgenesandsignalingpathwaysregulatedbytgfbr3invitro
AT seidmanjg transcriptionalprofilingofculturedembryonicepicardialcellsidentifiesnovelgenesandsignalingpathwaysregulatedbytgfbr3invitro
AT barnettjoeyv transcriptionalprofilingofculturedembryonicepicardialcellsidentifiesnovelgenesandsignalingpathwaysregulatedbytgfbr3invitro