Show simple item record

dc.contributor.authorMoreno Cubero, Elia 
dc.contributor.authorSubirá, Dolores
dc.contributor.authorSanz de Villalobos, Eduardo
dc.contributor.authorParra Cid, Trinidad
dc.contributor.authorMadejón, Antonio
dc.contributor.authorMiquel Plaza, Joaquín
dc.contributor.authorOlveira, Antonio
dc.contributor.authorGonzález Praetorius, Alejandro
dc.contributor.authorGarcía Samaniego, Javier
dc.contributor.authorLarrubia Marfil, Juan Ramón 
dc.date.accessioned2018-03-23T18:01:54Z
dc.date.issued2018-01-15
dc.identifier.bibliographicCitationJournal of virology, 2018, v. 92, n. 2, p. 1-23en
dc.identifier.urihttp://hdl.handle.net/10017/32839
dc.description.abstractHepatitis C virus (HCV)-specific CD8+T cells suffer a progressive exhaustion during persistent infection (PI) with HCV. This process could involve the positive immune checkpoint 4-1BB/4-1BBL through the loss of its signal transducer, TRAF1. To address this issue, peripheral HCV-specific CD8+T cells (pentamer-positive [pentamer+]/CD8+T cells) from patients with PI and resolved infection (RI) after treatment were studied. The duration of HCV infection and the liver fibrosis progression rate inversely correlated with the likelihood of detection of peripheral pentamer+/CD8+cells. In PI, pentamer+/CD8+cells had impaired antigen-specific reactivity that worsened when these cells were not detectableex vivoShort/midduration PI was characterized by detectable peripheral PD-1+CD127lowTRAF1lowcells. After triggering of T cell receptors (TCR), the TRAF1 level positively correlated with the levels of CD127, Mcl-1, and CD107a expression and proliferation intensity but negatively with PD-1 expression, linking TRAF1lowto exhaustion.In vitrotreatment with interleukin-7 (IL-7) upregulated TRAF1 expression, while treatment with transforming growth factor-β1 (TGF-β1) did the opposite, suggesting that the IL-7/TGF-β1 balance, besides TCR stimulation, could be involved in TRAF1 regulation. In fact, the serum TGF-β1 concentration was higher in patients with PI than in patients with RI, and it negatively correlated with TRAF1 expression. In line with IL-7 increasing the level of TRAF1 expression, IL-7 plus 4-1BBL treatmentin vitroenhanced T cell reactivity in patients with short/midduration infection. However, in patients with long-lasting PI, anti-PD-L1, in addition to the combination of IL-7 and 4-1BBL, was necessary to reestablish T cell proliferation in individuals with slowly progressing liver fibrosis (slow fibrosers) but had no effect in rapid fibrosers. In conclusion, a peripheral hyporeactive TRAF1lowHCV-specific CD8+T cell response, restorable by IL-7 plus 4-1BBL treatment, characterizes short/midduration PI. In long-lasting disease, HCV-specific CD8+T cells are rarely detectableex vivo, but treatment with IL-7, 4-1BBL, and anti-PD-L1 recovers their reactivityin vitroin slow fibrosers.IMPORTANCEHepatitis C virus (HCV) infects 71 million people worldwide. Two-thirds develop a chronic disease that can lead to cirrhosis and hepatocellular carcinoma. Direct-acting antivirals clear the infection, but there are still patients who relapse. In these cases, additional immunotherapy could play a vital role. A successful anti-HCV immune response depends on virus-specific CD8+T cells. During chronic infection, these cells are functionally impaired, which could be due to the failure of costimulation. This study describes exhausted specific T cells, characterized by low levels of expression of the signal transducer TRAF1 of the positive costimulatory pathway 4-1BB/4-1BBL. IL-7 upregulated TRAF1 expression and improved T cell reactivity in patients with short/midduration disease, while in patients with long-lasting infection, it was also necessary to block the negative PD-1/PD-L1 checkpoint. When the results are taken together, this work supports novel ways of restoring the specific CD8+T cell response, shedding light on the importance of TRAF1 signaling. This could be a promising target for future immunotherapy.en
dc.description.sponsorshipMinisterio de Economía y Competitividades_ES
dc.format.mimetypeapplication/pdfen
dc.language.isoengen
dc.publisherAmerican Society for Microbiologyen
dc.relation.hasparthttp://doi.org/10.21950/P2RKZB
dc.rightsAttribution-NonCommercial-NoDerivatives 4.0 Internacionalen
dc.rights.urihttp://creativecommons.org/licenses/by-nc-nd/4.0/en
dc.subjectHepatitis C virusen
dc.subjectImmune checkpointen
dc.subjectImmunotherapyen
dc.subjectCell exhaustionen
dc.titleAccording to Hepatitis C Virus (HCV) Infection Stage, Interleukin-7 Plus 4-1BB Triggering Alone or Combined with PD-1 Blockade Increases TRAF1lowHCV-Specific CD8+Cell Reactivity.en
dc.typeinfo:eu-repo/semantics/articleen
dc.subject.ecienciaMedicinaes_ES
dc.subject.ecienciaMedicineen
dc.contributor.affiliationUniversidad de Alcalá. Departamento de Medicina y Especialidades Médicas. Unidad docente Medicinaes_ES
dc.relation.projectIDinfo:eu-repo/grantAgreement/MINECO//PI12%2F00130/ESes_ES
dc.relation.projectIDinfo:eu-repo/grantAgreement/MINECO//PI15%2F00074/ES/Modulación del cluster miR 17-92 como mecanismo para restaurar la respuesta celular citotóxica específica contra antígenos asociados a carcinoma hepatocelulares_ES
dc.date.embargoEndDate2018-07-15
dc.rights.accessRightsinfo:eu-repo/semantics/openAccessen
dc.identifier.publicationtitleJournal of virologyen
dc.identifier.publicationvolume92
dc.identifier.publicationlastpage23
dc.identifier.publicationissue2
dc.identifier.publicationfirstpage1
dc.identifier.pmid29093082
dc.identifier.essn1098-5514


Files in this item

Thumbnail

This item appears in the following Collection(s)

Attribution-NonCommercial-NoDerivatives 4.0 Internacional
Este ítem está sujeto a una licencia Creative Commons.