Conserved gene microsynteny unveils functional interaction between protein disulfide isomerase and Rho Guanine-Dissociation Inhibitor Families

Detalhes bibliográficos
Autor(a) principal: Moretti, Ana I. S.
Data de Publicação: 2017
Outros Autores: Pavanelli, Jessyca C., Nolasco, Patrícia, Leisegang, Mathias S., Tanaka, Leonardo Y., Fernandes, Carolina Gonçalves, Wosniak Jr., João, Kajihara, Daniela, Dias, Matheus H., Fernandes, Denise C., Jo, Hanjoong, Tran, Neoc-Vinh, Ebersberger, Ingo, Brandes, Ralf P., Bonatto, Diego, Laurindo, Francisco R. M.
Tipo de documento: Artigo
Idioma: eng
Título da fonte: Repositório Institucional da UFRGS
Texto Completo: http://hdl.handle.net/10183/225551
Resumo: Protein disulfde isomerases (PDIs) support endoplasmic reticulum redox protein folding and cellsurface thiol-redox control of thrombosis and vascular remodeling. The family prototype PDIA1 regulates NADPH oxidase signaling and cytoskeleton organization, however the related underlying mechanisms are unclear. Here we show that genes encoding human PDIA1 and its two paralogs PDIA8 and PDIA2 are each fanked by genes encoding Rho guanine-dissociation inhibitors (GDI), known regulators of RhoGTPases/cytoskeleton. Evolutionary histories of these three microsyntenic regions reveal their emergence by two successive duplication events of a primordial gene pair in the last common vertebrate ancestor. The arrangement, however, is substantially older, detectable in echinoderms, nematodes, and cnidarians. Thus, PDI/RhoGDI pairing in the same transcription orientation emerged early in animal evolution and has been largely maintained. PDI/RhoGDI pairs are embedded into conserved genomic regions displaying common cis-regulatory elements. Analysis of gene expression datasets supports evidence for PDI/RhoGDI coexpression in developmental/ infammatory contexts. PDIA1/RhoGDIα were co-induced in endothelial cells upon CRISP-R-promoted transcription activation of each pair component, and also in mouse arterial intima during fow-induced remodeling. We provide evidence for physical interaction between both proteins. These data support strong functional links between PDI and RhoGDI families, which likely maintained PDI/RhoGDI microsynteny along>800-million years of evolution.
id UFRGS-2_1105179dd9630374e8a2c8dc329fecd0
oai_identifier_str oai:www.lume.ufrgs.br:10183/225551
network_acronym_str UFRGS-2
network_name_str Repositório Institucional da UFRGS
repository_id_str
spelling Moretti, Ana I. S.Pavanelli, Jessyca C.Nolasco, PatríciaLeisegang, Mathias S.Tanaka, Leonardo Y.Fernandes, Carolina GonçalvesWosniak Jr., JoãoKajihara, DanielaDias, Matheus H.Fernandes, Denise C.Jo, HanjoongTran, Neoc-VinhEbersberger, IngoBrandes, Ralf P.Bonatto, DiegoLaurindo, Francisco R. M.2021-08-11T04:48:30Z20172045-2322http://hdl.handle.net/10183/225551001070780Protein disulfde isomerases (PDIs) support endoplasmic reticulum redox protein folding and cellsurface thiol-redox control of thrombosis and vascular remodeling. The family prototype PDIA1 regulates NADPH oxidase signaling and cytoskeleton organization, however the related underlying mechanisms are unclear. Here we show that genes encoding human PDIA1 and its two paralogs PDIA8 and PDIA2 are each fanked by genes encoding Rho guanine-dissociation inhibitors (GDI), known regulators of RhoGTPases/cytoskeleton. Evolutionary histories of these three microsyntenic regions reveal their emergence by two successive duplication events of a primordial gene pair in the last common vertebrate ancestor. The arrangement, however, is substantially older, detectable in echinoderms, nematodes, and cnidarians. Thus, PDI/RhoGDI pairing in the same transcription orientation emerged early in animal evolution and has been largely maintained. PDI/RhoGDI pairs are embedded into conserved genomic regions displaying common cis-regulatory elements. Analysis of gene expression datasets supports evidence for PDI/RhoGDI coexpression in developmental/ infammatory contexts. PDIA1/RhoGDIα were co-induced in endothelial cells upon CRISP-R-promoted transcription activation of each pair component, and also in mouse arterial intima during fow-induced remodeling. We provide evidence for physical interaction between both proteins. These data support strong functional links between PDI and RhoGDI families, which likely maintained PDI/RhoGDI microsynteny along>800-million years of evolution.application/pdfengScientific reports. London. Vol. 7, (Dec. 2017), 17262, 1-18 p.Inibidores de dissulfeto isomeraseInibidores de guanina dissódicaConserved gene microsynteny unveils functional interaction between protein disulfide isomerase and Rho Guanine-Dissociation Inhibitor FamiliesEstrangeiroinfo:eu-repo/semantics/articleinfo:eu-repo/semantics/publishedVersioninfo:eu-repo/semantics/openAccessreponame:Repositório Institucional da UFRGSinstname:Universidade Federal do Rio Grande do Sul (UFRGS)instacron:UFRGSTEXT001070780.pdf.txt001070780.pdf.txtExtracted Texttext/plain89741http://www.lume.ufrgs.br/bitstream/10183/225551/2/001070780.pdf.txte7f9e4be4dc7effeb61febab4b556ebeMD52ORIGINAL001070780.pdfTexto completo (inglês)application/pdf5042982http://www.lume.ufrgs.br/bitstream/10183/225551/1/001070780.pdf2d88747b2400285fec5b7fb2b61e7c0eMD5110183/2255512021-08-18 04:46:38.626762oai:www.lume.ufrgs.br:10183/225551Repositório de PublicaçõesPUBhttps://lume.ufrgs.br/oai/requestopendoar:2021-08-18T07:46:38Repositório Institucional da UFRGS - Universidade Federal do Rio Grande do Sul (UFRGS)false
dc.title.pt_BR.fl_str_mv Conserved gene microsynteny unveils functional interaction between protein disulfide isomerase and Rho Guanine-Dissociation Inhibitor Families
title Conserved gene microsynteny unveils functional interaction between protein disulfide isomerase and Rho Guanine-Dissociation Inhibitor Families
spellingShingle Conserved gene microsynteny unveils functional interaction between protein disulfide isomerase and Rho Guanine-Dissociation Inhibitor Families
Moretti, Ana I. S.
Inibidores de dissulfeto isomerase
Inibidores de guanina dissódica
title_short Conserved gene microsynteny unveils functional interaction between protein disulfide isomerase and Rho Guanine-Dissociation Inhibitor Families
title_full Conserved gene microsynteny unveils functional interaction between protein disulfide isomerase and Rho Guanine-Dissociation Inhibitor Families
title_fullStr Conserved gene microsynteny unveils functional interaction between protein disulfide isomerase and Rho Guanine-Dissociation Inhibitor Families
title_full_unstemmed Conserved gene microsynteny unveils functional interaction between protein disulfide isomerase and Rho Guanine-Dissociation Inhibitor Families
title_sort Conserved gene microsynteny unveils functional interaction between protein disulfide isomerase and Rho Guanine-Dissociation Inhibitor Families
author Moretti, Ana I. S.
author_facet Moretti, Ana I. S.
Pavanelli, Jessyca C.
Nolasco, Patrícia
Leisegang, Mathias S.
Tanaka, Leonardo Y.
Fernandes, Carolina Gonçalves
Wosniak Jr., João
Kajihara, Daniela
Dias, Matheus H.
Fernandes, Denise C.
Jo, Hanjoong
Tran, Neoc-Vinh
Ebersberger, Ingo
Brandes, Ralf P.
Bonatto, Diego
Laurindo, Francisco R. M.
author_role author
author2 Pavanelli, Jessyca C.
Nolasco, Patrícia
Leisegang, Mathias S.
Tanaka, Leonardo Y.
Fernandes, Carolina Gonçalves
Wosniak Jr., João
Kajihara, Daniela
Dias, Matheus H.
Fernandes, Denise C.
Jo, Hanjoong
Tran, Neoc-Vinh
Ebersberger, Ingo
Brandes, Ralf P.
Bonatto, Diego
Laurindo, Francisco R. M.
author2_role author
author
author
author
author
author
author
author
author
author
author
author
author
author
author
dc.contributor.author.fl_str_mv Moretti, Ana I. S.
Pavanelli, Jessyca C.
Nolasco, Patrícia
Leisegang, Mathias S.
Tanaka, Leonardo Y.
Fernandes, Carolina Gonçalves
Wosniak Jr., João
Kajihara, Daniela
Dias, Matheus H.
Fernandes, Denise C.
Jo, Hanjoong
Tran, Neoc-Vinh
Ebersberger, Ingo
Brandes, Ralf P.
Bonatto, Diego
Laurindo, Francisco R. M.
dc.subject.por.fl_str_mv Inibidores de dissulfeto isomerase
Inibidores de guanina dissódica
topic Inibidores de dissulfeto isomerase
Inibidores de guanina dissódica
description Protein disulfde isomerases (PDIs) support endoplasmic reticulum redox protein folding and cellsurface thiol-redox control of thrombosis and vascular remodeling. The family prototype PDIA1 regulates NADPH oxidase signaling and cytoskeleton organization, however the related underlying mechanisms are unclear. Here we show that genes encoding human PDIA1 and its two paralogs PDIA8 and PDIA2 are each fanked by genes encoding Rho guanine-dissociation inhibitors (GDI), known regulators of RhoGTPases/cytoskeleton. Evolutionary histories of these three microsyntenic regions reveal their emergence by two successive duplication events of a primordial gene pair in the last common vertebrate ancestor. The arrangement, however, is substantially older, detectable in echinoderms, nematodes, and cnidarians. Thus, PDI/RhoGDI pairing in the same transcription orientation emerged early in animal evolution and has been largely maintained. PDI/RhoGDI pairs are embedded into conserved genomic regions displaying common cis-regulatory elements. Analysis of gene expression datasets supports evidence for PDI/RhoGDI coexpression in developmental/ infammatory contexts. PDIA1/RhoGDIα were co-induced in endothelial cells upon CRISP-R-promoted transcription activation of each pair component, and also in mouse arterial intima during fow-induced remodeling. We provide evidence for physical interaction between both proteins. These data support strong functional links between PDI and RhoGDI families, which likely maintained PDI/RhoGDI microsynteny along>800-million years of evolution.
publishDate 2017
dc.date.issued.fl_str_mv 2017
dc.date.accessioned.fl_str_mv 2021-08-11T04:48:30Z
dc.type.driver.fl_str_mv Estrangeiro
info:eu-repo/semantics/article
dc.type.status.fl_str_mv info:eu-repo/semantics/publishedVersion
format article
status_str publishedVersion
dc.identifier.uri.fl_str_mv http://hdl.handle.net/10183/225551
dc.identifier.issn.pt_BR.fl_str_mv 2045-2322
dc.identifier.nrb.pt_BR.fl_str_mv 001070780
identifier_str_mv 2045-2322
001070780
url http://hdl.handle.net/10183/225551
dc.language.iso.fl_str_mv eng
language eng
dc.relation.ispartof.pt_BR.fl_str_mv Scientific reports. London. Vol. 7, (Dec. 2017), 17262, 1-18 p.
dc.rights.driver.fl_str_mv info:eu-repo/semantics/openAccess
eu_rights_str_mv openAccess
dc.format.none.fl_str_mv application/pdf
dc.source.none.fl_str_mv reponame:Repositório Institucional da UFRGS
instname:Universidade Federal do Rio Grande do Sul (UFRGS)
instacron:UFRGS
instname_str Universidade Federal do Rio Grande do Sul (UFRGS)
instacron_str UFRGS
institution UFRGS
reponame_str Repositório Institucional da UFRGS
collection Repositório Institucional da UFRGS
bitstream.url.fl_str_mv http://www.lume.ufrgs.br/bitstream/10183/225551/2/001070780.pdf.txt
http://www.lume.ufrgs.br/bitstream/10183/225551/1/001070780.pdf
bitstream.checksum.fl_str_mv e7f9e4be4dc7effeb61febab4b556ebe
2d88747b2400285fec5b7fb2b61e7c0e
bitstream.checksumAlgorithm.fl_str_mv MD5
MD5
repository.name.fl_str_mv Repositório Institucional da UFRGS - Universidade Federal do Rio Grande do Sul (UFRGS)
repository.mail.fl_str_mv
_version_ 1801225032125382656