Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Front Immunol ; 13: 918928, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35812370

RESUMO

To provide a unique global view of the relative potential for evasion of CD8+ and CD4+ T cells by SARS-CoV-2 lineages as they evolve over time, we performed a comprehensive analysis of predicted HLA-I and HLA-II binding peptides in Spike (S) and Nucleocapsid (N) protein sequences of all available SARS-CoV-2 genomes as provided by NIH NCBI at a bi-monthly interval between March and December of 2021. A data supplement of all B.1.1.529 (Omicron) genomes from GISAID in early December was also used to capture the rapidly spreading variant. A key finding is that throughout continued viral evolution and increasing rates of mutations occurring at T-cell epitope hotspots, protein instances with worst-case binding loss did not become the most frequent for any Variant of Concern (VOC) or Variant of Interest (VOI) lineage; suggesting T-cell evasion is not likely to be a dominant evolutionary pressure on SARS-CoV-2. We also determined that throughout the course of the pandemic in 2021, there remained a relatively steady ratio of viral variants that exhibit conservation of epitopes in the N protein, despite significant potential for epitope loss in S relative to other lineages. We further localized conserved regions in N with high epitope yield potential, and illustrated heterogeneity in HLA-I binding across the S protein consistent with empirical observations. Although Omicron's high volume of mutations caused it to exhibit more epitope loss potential than most frequently observed versions of proteins in almost all other VOCs, epitope candidates across its most frequent N proteins were still largely conserved. This analysis adds to the body of evidence suggesting that N may have merit as an additional antigen to elicit immune responses to vaccination with increased potential to provide sustained protection against COVID-19 disease in the face of emerging variants.


Assuntos
COVID-19 , SARS-CoV-2 , COVID-19/genética , Epitopos de Linfócito T , Humanos , Nucleocapsídeo/metabolismo , SARS-CoV-2/genética , Glicoproteína da Espícula de Coronavírus
2.
iScience ; 20: 119-136, 2019 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-31563852

RESUMO

DNA accessibility is a key dynamic feature of chromatin regulation that can potentiate transcriptional events and tumor progression. To gain insight into chromatin state across existing tumor data, we improved neural network models for predicting accessibility from DNA sequence and extended them to incorporate a global set of RNA sequencing gene expression inputs. Our expression-informed model expanded the application domain beyond specific tissue types to tissues not present in training and achieved consistently high accuracy in predicting DNA accessibility at promoter and promoter flank regions. We then leveraged our new tool by analyzing the DNA accessibility landscape of promoters across The Cancer Genome Atlas. We show that in lung adenocarcinoma the accessibility perspective uniquely highlights immune pathways inversely correlated with a more open chromatin state and that accessibility patterns learned from even a single tumor type can discriminate immune inflammation across many cancers, often with direct relation to patient prognosis.

3.
NPJ Genom Med ; 3: 14, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29928512

RESUMO

Immune heterogeneity within the tumor microenvironment undoubtedly adds several layers of complexity to our understanding of drug sensitivity and patient prognosis across various cancer types. Within the tumor microenvironment, immunogenicity is a favorable clinical feature in part driven by the antitumor activity of CD8+ T cells. However, tumors often inhibit this antitumor activity by exploiting the suppressive function of regulatory T cells (Tregs), thus suppressing the adaptive immune response. Despite the seemingly intuitive immunosuppressive biology of Tregs, prognostic studies have produced contradictory results regarding the relationship between Treg enrichment and survival. We therefore analyzed RNA-seq data of Treg-enriched tumor samples to derive a pan-cancer gene signature able to help reconcile the inconsistent results of Treg studies, by better understanding the variable clinical association of Tregs across alternative tumor contexts. We show that increased expression of a 32-gene signature in Treg-enriched tumor samples (n = 135) is able to distinguish a cohort of patients associated with chemosensitivity and overall survival. This cohort is also enriched for CD8+ T cell abundance, as well as the antitumor M1 macrophage subtype. With a subsequent validation in a larger TCGA pool of Treg-enriched patients (n = 626), our results reveal a gene signature able to produce unsupervised clusters of Treg-enriched patients, with one cluster of patients uniquely representative of an immunogenic tumor microenvironment. Ultimately, these results support the proposed gene signature as a putative biomarker to identify certain Treg-enriched patients with immunogenic tumors that are more likely to be associated with features of favorable clinical outcome.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA