Skip navigation
Veuillez utiliser cette adresse pour citer ce document : http://repositorio2.unb.br/jspui/handle/10482/43672
Fichier(s) constituant ce document :
Fichier Description TailleFormat 
ARTIGO_InflammatoryStatusSoluble.pdf2,73 MBAdobe PDFVoir/Ouvrir
Affichage complet
Élément Dublin CoreValeurLangue
dc.contributor.authorBogéa, Gabriela Muller Reche-
dc.contributor.authorCarvalho, Amandda Évelin Silva de-
dc.contributor.authorBraga, Luma Dayane de Carvalho Filiú-
dc.contributor.authorNeves, Francisco de Assis Rocha-
dc.contributor.authorAraujo, Felipe Saldanha de-
dc.date.accessioned2022-05-09T13:21:34Z-
dc.date.available2022-05-09T13:21:34Z-
dc.date.issued2022-
dc.identifier.citationBOGÉA, Gabriela Muller Reche et al. The inflammatory status of soluble microenvironment influences the capacity of melanoma cells to control t-cell responses. Frontiers in Oncology, v. 12, 858425, 2022. DOI: https://doi.org/10.3389/fonc.2022.858425. Disponível em: https://www.frontiersin.org/articles/10.3389/fonc.2022.858425/full. Acesso em: 9 maio 2022.pt_BR
dc.identifier.urihttps://repositorio.unb.br/handle/10482/43672-
dc.language.isoInglêspt_BR
dc.publisherFrontierspt_BR
dc.rightsAcesso Abertopt_BR
dc.titleThe inflammatory status of soluble microenvironment influences the capacity of melanoma cells to control t-cell responsespt_BR
dc.typeArtigopt_BR
dc.subject.keywordMelanomapt_BR
dc.subject.keywordCélulas Tpt_BR
dc.subject.keywordResposta imunológicapt_BR
dc.subject.keywordSecretomapt_BR
dc.rights.license(CC BY)pt_BR
dc.identifier.doihttps://doi.org/10.3389/fonc.2022.858425pt_BR
dc.description.abstract1The development of immunotherapeutic approaches for the treatment of melanoma requires a better understanding of immunoescape mechanisms of tumor cells and how they interact with other tumor-resident cell types. Here, we evaluated how the conditioned media of resting (rCM) and immune-activated PBMCs (iCM) influence the ability of a metastatic melanoma cell line (MeWo) to control T-cells function. MeWo cells were expanded in RPMI, rCM, or iCM and the secretome generated after cell expansion was identified as MeSec (RPMI), niSec (non-inflammatory), or iSec (inflammatory secretome), respectively. Then, the immunomodulatory potential of such secretomes was tested in PHA-activated PBMCs. iCM induced higher levels of IFN-γ and IL-10 in treated melanoma cells compared to rCM, as well as higher IDO and PD-L1 expression. The iSec was able to inhibit T-cell activation and proliferation. Interestingly, PBMCs treated with iSec presented a reduced expression of the regulators of Th1 and Th2 responses T-BET and GATA-3, as well as low expression of IFN-γ, and co-stimulatory molecules TIM-3 and LAG-3. Importantly, our findings show that melanoma may benefit from an inflammatory microenvironment to enhance its ability to control the T-cell response. Interestingly, such an immunomodulatory effect involves the inhibition of the checkpoint molecules LAG-3 and TIM-3, which are currently investigated as important therapeutic targets for melanoma treatment. Further studies are needed to better understand how checkpoint molecules are modulated by paracrine and cell contact-dependent interaction between melanoma and immune cells. Such advances are fundamental for the development of new therapeutic approaches focused on melanoma immunotherapy.pt_BR
Collection(s) :Artigos publicados em periódicos e afins

Affichage abbrégé " class="statisticsLink btn btn-primary" href="/jspui/handle/10482/43672/statistics">



Tous les documents dans DSpace sont protégés par copyright, avec tous droits réservés.