Please use this identifier to cite or link to this item: https://ninho.inca.gov.br/jspui/handle/123456789/6188
Full metadata record
DC FieldValueLanguage
dc.contributor.advisorO-GlcNAcaseen
dc.contributor.authorJulião, Glaucia-
dc.contributor.authorMenezes, Aline-
dc.contributor.authorMariath, Fernanda-
dc.contributor.authorHanover, John A.-
dc.contributor.authorEvaristo, Joseph Albert Medeiros-
dc.contributor.authorNogueira, Fábio César Sousa-
dc.contributor.authorDias, Wagner Barbosa-
dc.contributor.authorPereira, Denise de Abreu-
dc.contributor.authorCarneiro, Katia-
dc.contributor.authorNunes, Maria Cecília Oliveira Ferreira-
dc.date.accessioned2022-04-01T15:17:55Z-
dc.date.available2022-04-01T15:17:55Z-
dc.date.issued2021-04-
dc.identifier.issn1542-6416-
dc.identifier.other10.1186/s12014‑021‑09317‑x-
dc.identifier.urihttp://sr-vmlxaph03:8080/jspui/handle/123456789/6188-
dc.description.abstractGlioblastoma (GBM) is a grade IV glioma highly aggressive and refractory to the therapeutic approaches currently in use. O‑GlcNAcylation plays a key role for tumor aggressiveness and progression in different types of cancer; however, experimental evidence of its involvement in GBM are still lacking. Here, we show that O‑GlcNAcylation plays a critical role in maintaining the composition of the GBM secretome, whereas inhibition of OGA activity disrupts the intercel‑ lular signaling via microvesicles. Using a label‑free quantitative proteomics methodology, we identified 51 proteins in the GBM secretome whose abundance was significantly altered by activity inhibition of O‑GlcNAcase (iOGA). Among these proteins, we observed that proteins related to proteasome activity and to regulation of immune response in the tumor microenvironment were consistently downregulated in GBM cells upon iOGA. While the proteins IGFBP3, IL‑6 and HSPA5 were downregulated in GBM iOGA cells, the protein SQSTM1/p62 was exclusively found in GBM cells under iOGA. These findings were in line with literature evidence on the role of p62/IL‑6 signaling axis in suppressing tumor aggressiveness and our experimental evidence showing a decrease in radioresistance potential of these cells. Taken together, our findings provide evidence that OGA activity may regulate the p62 and IL‑6 abundance in the GBM secretome. We propose that the assessment of tumor status from the main proteins present in its secretome may contribute to the advancement of diagnostic, prognostic and even therapeutic tools to approach this relevant malignancy.pt_BR
dc.publisherClinical Proteomicspt_BR
dc.subjectGlioblastomapt_BR
dc.subject.otherSecretomeen
dc.subject.otherO-GlcNAcylationen
dc.titleO‑GlcNAcylation protein disruption by Thiamet G promotes changes on the GBM U87‑MG cells secretome molecular signaturept_BR
dc.TypeArticlept_BR
Appears in Collections:Artigos de Periódicos da Pesquisa Experimental e Translacional



Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.