Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 14: 1173035, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37197667

RESUMO

Cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) is an immune checkpoint expressed in regulatory T (Treg) cells and activated T lymphocytes. Despite its potential as a treatment strategy for melanoma, CTLA-4 inhibition has limited efficacy. Using data from The Cancer Genome Atlas (TCGA) melanoma database and another dataset, we found that decreased CTLA4 mRNA was associated with a poorer prognosis in metastatic melanoma. To investigate further, we measured blood CTLA4 mRNA in 273 whole-blood samples from an Australian cohort and found that it was lower in metastatic melanoma than in healthy controls and associated with worse patient survival. We confirmed these findings using Cox proportional hazards model analysis and another cohort from the US. Fractionated blood analysis revealed that Treg cells were responsible for the downregulated CTLA4 in metastatic melanoma patients, which was confirmed by further analysis of published data showing downregulated CTLA-4 surface protein expression in Treg cells of metastatic melanoma compared to healthy donors. Mechanistically, we found that secretomes from human metastatic melanoma cells downregulate CTLA4 mRNA at the post-transcriptional level through miR-155 while upregulating FOXP3 expression in human Treg cells. Functionally, we demonstrated that CTLA4 expression inhibits the proliferation and suppressive function of human Treg cells. Finally, miR-155 was found to be upregulated in Treg cells from metastatic melanoma patients compared to healthy donors. Our study provides new insights into the underlying mechanisms of reduced CTLA4 expression observed in melanoma patients, demonstrating that post-transcriptional silencing of CTLA4 by miRNA-155 in Treg cells may play a critical role. Since CTLA-4 expression is downregulated in non-responder melanoma patients to anti-PD-1 immunotherapy, targeting miRNA-155 or other factors involved in regulating CTLA4 expression in Treg cells without affecting T cells could be a potential strategy to improve the efficacy of immunotherapy in melanoma. Further research is needed to understand the molecular mechanisms regulating CTLA4 expression in Treg cells and identify potential therapeutic targets for enhancing immune-based therapies.


Assuntos
Melanoma , MicroRNAs , Segunda Neoplasia Primária , Humanos , Linfócitos T Reguladores , Antígeno CTLA-4 , Austrália , Prognóstico , MicroRNAs/metabolismo
3.
Oncotarget ; 10(27): 2583-2585, 2019 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-31080551

RESUMO

The concept of cancer stemness has undergone a paradigm shift during the last decade where there is wider acceptance of the idea that stemness in cancer is a more dynamic and plastic phenomenon than previously thought. However, we have yet to understand the mechanisms on how this stochastic plasticity arises and is maintained. Recently, we have shown that CDK1 plays a critical role in stochastic stemness and tumor initiation potential through regulating SOX2 phosphorylation in multiple cancer types. The phosphorylation of SOX2 affects its nuclear localization, thereby determining the transcriptional fate of its downstream targets. We have also validated the significance of these findings using clinical samples by demonstrating that CDK1high tumor samples displayed upregulation of MYC target genes, which were reported to overlap with SOX2 targets. In the current article, we further discuss the possibility of a closed, feed-forward loop between SOX2 and CDK1 through a long non-coding RNA, CCAT1, which would explain the sustained activation of this loop. Despite the extensive investigation of the cancer stemness as a cause of drug resistance, its role in immune evasion still requires further understanding, and hence, in this article, we further discuss the possibility of this CDK1-SOX2 axis contributing to immune resistance through modulating cell-to-cell interaction directly or indirectly in the tumor microenvironment.

4.
Oncotarget ; 9(9): 8206-8222, 2018 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-29492189

RESUMO

Besides somatic mutations or drug efflux, epigenetic reprogramming can lead to acquired drug resistance. We recently have identified early stress-induced multi-drug tolerant cancer cells termed induced drug-tolerant cells (IDTCs). Here, IDTCs were generated using different types of cancer cell lines; melanoma, lung, breast and colon cancer. A common loss of the H3K4me3 and H3K27me3 and gain of H3K9me3 mark was observed as a significant response to drug exposure or nutrient starvation in IDTCs. These epigenetic changes were reversible upon drug holidays. Microarray, qRT-PCR and protein expression data confirmed the up-regulation of histone methyltransferases (SETDB1 and SETDB2) which contribute to the accumulation of H3K9me3 concomitantly in the different cancer types. Genome-wide studies suggest that transcriptional repression of genes is due to concordant loss of H3K4me3 and regional increment of H3K9me3. Conversely, genome-wide CpG site-specific DNA methylation showed no common changes at the IDTC state. This suggests that distinct histone methylation patterns rather than DNA methylation are driving the transition from parental to IDTCs. In addition, silencing of SETDB1/2 reversed multi drug tolerance. Alterations of histone marks in early multi-drug tolerance with an increment in H3K9me3 and loss of H3K4me3/H3K27me3 is neither exclusive for any particular stress response nor cancer type specific but rather a generic response.

5.
J Invest Dermatol ; 135(12): 2923-2925, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26569587

RESUMO

Increasingly comprehensive observations indicate that the tumor microenvironment contributes to drug resistance toward small molecule inhibitors. Fedorenko et al. describe a role for fibroblasts in creating a favorable niche for melanoma cell survival if treated with the BRAF inhibitor vemurafenib. TGF-ß released by vemurafenib-treated melanoma cells stimulated fibroblasts for increased α-smooth muscle actin, neuregulin (NRG), and fibronectin expression. Off-target effects of vemurafenib led to paradoxical secretion of hepatocyte growth factor (HGF) by fibroblasts. Combined inhibition of BRAF/MET/HER kinases was insufficient to reverse the protective effect of the fibroblasts, whereas reversal was achieved by combined BRAF/PI3K inhibition. A thorough understanding of the complex spatiotemporal interactions in tumor microenvironments holds promise for improved targeting using combination therapies in patients with melanoma.


Assuntos
Melanoma/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...