Identificador persistente para citar o vincular este elemento: http://hdl.handle.net/10553/117369
Campo DC Valoridioma
dc.contributor.authorFernández-García, Victoriaen_US
dc.contributor.authorGonzález-Ramos, Silviaen_US
dc.contributor.authorAvendaño-Ortiz, Joséen_US
dc.contributor.authorMartín-Sanz, Palomaen_US
dc.contributor.authorGómez-Coronado, Diegoen_US
dc.contributor.authorDelgado, Carmenen_US
dc.contributor.authorCastrillo, Antonioen_US
dc.contributor.authorBoscá, Lisardoen_US
dc.date.accessioned2022-07-26T11:52:22Z-
dc.date.available2022-07-26T11:52:22Z-
dc.date.issued2022en_US
dc.identifier.issn1420-682Xen_US
dc.identifier.otherScopus-
dc.identifier.otherWoS-
dc.identifier.urihttp://hdl.handle.net/10553/117369-
dc.description.abstractIn the course of atherogenesis, the spleen plays an important role in the regulation of extramedullary hematopoiesis, and in the control of circulating immune cells, which contributes to plaque progression. Here, we have investigated the role of splenic nucleotide-binding oligomerization domain 1 (NOD1) in the recruitment of circulating immune cells, as well as the involvement of this immune organ in extramedullary hematopoiesis in mice fed on a high-fat high-cholesterol diet (HFD). Under HFD conditions, the absence of NOD1 enhances the mobilization of immune cells, mainly neutrophils, from the bone marrow to the blood. To determine the effect of NOD1-dependent mobilization of immune cells under pro-atherogenic conditions, Apoe−/− and Apoe−/−Nod1−/− mice fed on HFD for 4 weeks were used. Splenic NOD1 from Apoe−/− mice was activated after feeding HFD as inferred by the phosphorylation of the NOD1 downstream targets RIPK2 and TAK1. Moreover, this activation was accompanied by the release of neutrophil extracellular traps (NETs), as determined by the increase in the expression of peptidyl arginine deiminase 4, and the identification of citrullinated histone H3 in this organ. This formation of NETs was significantly reduced in Apoe−/−Nod1−/− mice. Indeed, the presence of Ly6G+ cells and the lipidic content in the spleen of mice deficient in Apoe and Nod1 was reduced when compared to the Apoe−/− counterparts, which suggests that the mobilization and activation of circulating immune cells are altered in the absence of NOD1. Furthermore, confirming previous studies, Apoe−/−Nod1−/− mice showed a reduced atherogenic disease, and diminished recruitment of neutrophils in the spleen, compared to Apoe−/− mice. However, splenic artery ligation reduced the atherogenic burden in Apoe−/− mice an effect that, unexpectedly was lost in Apoe−/−Nod1−/− mice. Together, these results suggest that neutrophil accumulation and activity in the spleen are driven in part by NOD1 activation in mice fed on HFD, contributing in this way to regulating atherogenic progression.en_US
dc.languageengen_US
dc.relation.ispartofCellular and Molecular Life Sciencesen_US
dc.sourceCellular and Molecular Life Sciences [ISSN 1420-682X], v. 79 (8), 396, (Agosto 2022)en_US
dc.subject32 Ciencias médicasen_US
dc.subject2407 Biología celularen_US
dc.subject2403 Bioquímicaen_US
dc.subject320502 Endocrinologíaen_US
dc.subject.otherAtherogenesisen_US
dc.subject.otherHypercholesterolemiaen_US
dc.subject.otherNETosisen_US
dc.subject.otherPartial Splenectomyen_US
dc.subject.otherSpleenen_US
dc.titleHigh-fat diet activates splenic NOD1 and enhances neutrophil recruitment and neutrophil extracellular traps release in the spleen of ApoE-deficient miceen_US
dc.typeinfo:eu-repo/semantics/Articleen_US
dc.typeArticleen_US
dc.identifier.doi10.1007/s00018-022-04415-xen_US
dc.identifier.scopus85133219619-
dc.identifier.isi000821012300003-
dc.contributor.orcidNO DATA-
dc.contributor.orcidNO DATA-
dc.contributor.orcidNO DATA-
dc.contributor.orcidNO DATA-
dc.contributor.orcidNO DATA-
dc.contributor.orcidNO DATA-
dc.contributor.orcidNO DATA-
dc.contributor.orcid0000-0002-0253-5469-
dc.contributor.authorscopusid57201801330-
dc.contributor.authorscopusid56041607200-
dc.contributor.authorscopusid57163917300-
dc.contributor.authorscopusid57196054763-
dc.contributor.authorscopusid6603760125-
dc.contributor.authorscopusid55428853800-
dc.contributor.authorscopusid55445301000-
dc.contributor.authorscopusid35514045400-
dc.identifier.eissn1420-9071-
dc.identifier.issue8-
dc.relation.volume79en_US
dc.investigacionCiencias de la Saluden_US
dc.type2Artículoen_US
dc.contributor.daisngid15619456-
dc.contributor.daisngid29578949-
dc.contributor.daisngid15713237-
dc.contributor.daisngid11632080-
dc.contributor.daisngid15461631-
dc.contributor.daisngid6948351-
dc.contributor.daisngid5672318-
dc.contributor.daisngid29643912-
dc.description.numberofpages15en_US
dc.utils.revisionen_US
dc.contributor.wosstandardWOS:Fernandez-Garcia, V-
dc.contributor.wosstandardWOS:Gonzalez-Ramos, S-
dc.contributor.wosstandardWOS:Avendano-Ortiz, J-
dc.contributor.wosstandardWOS:Martin-Sanz, P-
dc.contributor.wosstandardWOS:Gomez-Coronado, D-
dc.contributor.wosstandardWOS:Delgado, C-
dc.contributor.wosstandardWOS:Castrillo, A-
dc.contributor.wosstandardWOS:Bosca, L-
dc.date.coverdateAgosto 2022en_US
dc.identifier.ulpgcen_US
dc.contributor.buulpgcBU-MEDen_US
dc.description.sjr2,371-
dc.description.jcr8,0-
dc.description.sjrqQ1-
dc.description.jcrqQ1-
dc.description.scieSCIE-
dc.description.miaricds10,9-
item.grantfulltextopen-
item.fulltextCon texto completo-
crisitem.author.deptGIR IUIBS: Farmacología Molecular y Traslacional-
crisitem.author.deptIU de Investigaciones Biomédicas y Sanitarias-
crisitem.author.orcid0000-0002-2057-2159-
crisitem.author.parentorgIU de Investigaciones Biomédicas y Sanitarias-
crisitem.author.fullNameCastrillo Viguera, Antonio Jesús-
Colección:Artículos
Adobe PDF (1,94 MB)
Vista resumida

Google ScholarTM

Verifica

Altmetric


Comparte



Exporta metadatos



Los elementos en ULPGC accedaCRIS están protegidos por derechos de autor con todos los derechos reservados, a menos que se indique lo contrario.