Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cells ; 9(2)2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-32028704

RESUMO

miR-28-5p is downregulated in some tumor tissues in which it has been demonstrated to have tumor suppressor (TS) activity. Here, we demonstrate that miR-28-5p acts as a TS in prostate cancer (PCa) cells affecting cell proliferation/survival, as well as migration and invasion. Using the miRNA pull out assay and next generation sequencing, we collected the complete repertoire of miR-28-5p targets, obtaining a data set (miR-28-5p targetome) of 191 mRNAs. Filtering the targetome with TargetScan 7, PITA and RNA22, we found that 61% of the transcripts had miR-28-5p binding sites. To assign a functional value to the captured transcripts, we grouped the miR-28-5p targets into gene families with annotated function and showed that six transcripts belong to the transcription factor category. Among them we selected SREBF2, a gene with an important role in PCa. We validated miR-28-5p/SREBF2 interaction, demonstrating that SREBF2 inhibition affects almost all the tumor processes altered by miR-28-5p re-expression, suggesting that SREBF2 is an important mediator of miR-28-5p TS activity. Our findings support the identification of the targetome of cancer-related miRNAs as a tool to discover genes and pathways fundamental for tumor development, and potential new targets for anti-tumor therapy.


Assuntos
Genes Supressores de Tumor , MicroRNAs/metabolismo , Neoplasias da Próstata/genética , Proteína de Ligação a Elemento Regulador de Esterol 2/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , MicroRNAs/genética , Proteína de Ligação a Elemento Regulador de Esterol 2/genética
2.
Biochim Biophys Acta Mol Basis Dis ; 1865(9): 2111-2124, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31004710

RESUMO

The capacity of inducing angiogenesis is a recognized hallmark of cancer cells. The cancer microenvironment, characterized by hypoxia and inflammatory signals, promotes proliferation, migration and activation of quiescent endothelial cells (EC) from surrounding vascular network. Current anti-angiogenic drugs present side effects, temporary efficacy, and issues of primary resistance, thereby calling for the identification of new therapeutic targets. MICALs are a unique family of redox enzymes that destabilize F-actin in cytoskeletal dynamics. MICAL2 mediates Semaphorin3A-NRP2 response to VEGFR1 in rat ECs. MICAL2 also enters the p130Cas interactome in response to VEGF in HUVEC. Previously, we showed that MICAL2 is overexpressed in metastatic cancer. A small-molecule inhibitor of MICAL2 exists (CCG-1423). Here we report that 1) MICAL2 is expressed in neo-angiogenic ECs in human solid tumors (kidney and breast carcinoma, glioblastoma and cardiac myxoma, n = 67, were analyzed with immunohistochemistry) and in animal models of ischemia/inflammation neo-angiogenesis, but not in normal capillary bed; 2) MICAL2 protein pharmacological inhibition (CCG-1423) or gene KD reduce EC viability and functional performance; 3) MICAL2 KD disables ECs response to VEGF in vitro. Whole-genome gene expression profiling reveals MICAL2 involvement in angiogenesis and vascular development pathways. Based on these results, we propose that MICAL2 expression in ECs participates to inflammation-induced neo-angiogenesis and that MICAL2 inhibition should be tested in cancer- and noncancer-associated neo-angiogenesis, where chronic inflammation represents a relevant pathophysiological mechanism.


Assuntos
Movimento Celular , Proteínas dos Microfilamentos/metabolismo , Oxirredutases/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Anilidas/farmacologia , Animais , Benzamidas/farmacologia , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células , Sobrevivência Celular/efeitos dos fármacos , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Expressão Gênica , Humanos , Masculino , Proteínas dos Microfilamentos/antagonistas & inibidores , Proteínas dos Microfilamentos/genética , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Neoplasias/irrigação sanguínea , Neoplasias/patologia , Neovascularização Patológica , Neovascularização Fisiológica , Oxirredutases/antagonistas & inibidores , Oxirredutases/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Wistar
3.
Int J Genomics ; 2017: 5214806, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29085832

RESUMO

miR-28-5p is an intragenic miRNA which is underexpressed in several tumor types showing a tumor suppressor (TS) activity. Routinely, the known miR-28-5p targets are validated in specific tumor contexts but it is unclear whether these targets are also being regulated in other tumor types. To this end, we adopted the miRNA pull out assay to capture the miR-28-5p targets in DU-145 prostate cancer (PCa) cells. Firstly, we demonstrated that miR-28-5p acts as a TS-miRNA in PCa, affecting cell proliferation, survival, and apoptosis. Secondly, we evaluated the enrichment of the 10 validated miR-28-5p targets in the pull out sample. We showed that E2F6, TEX-261, MAPK1, MPL, N4BP1, and RAP1B but not BAG1, OTUB1, MAD2L1, and p21 were significantly enriched, suggesting that not all the miR-28-5p targets are regulated by this miRNA in PCa. We then verified whether the miR-28-5p-interacting targets were regulated by this miRNA. We selected E2F6, the most enriched target in the pull out sample, and demonstrated that miR-28-5p downregulated E2F6 at the protein level suggesting that our approach was effective. In general terms, these findings support the miRNA pull out assay as a useful method to identify context-specific miRNA targets.

4.
J Cancer ; 8(14): 2729-2739, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28928862

RESUMO

Purpose. miR-26a-5p is a tumor suppressor (TS) miRNA often downregulated in several tumor tissues and tumor cell lines. In this work, we performed the re-expression of the miR-26a-5p in DU-145 prostate cancer cells to collect genes interacting with miR-26a-5p and analyzed their integration in the tumorigenesis related pathways. Methods. The transfection of DU-145 prostate cancer cells with miR-26a-5p was done using nucleofection. The biological effects induced by miR-26a-5p re-expression were detected with routine assays for cell proliferation, cell cycle, survival, apoptosis and cell migration. The miRNA pull out technique was used to collect and next generation sequencing to identify the complete repertoire of the miR-26a-5p targets (miR-26a-5p/targetome). TargetScan 7, PITA and RNA22 were used to find the predicted miR-26a-5p targets in the miR-26a-5p/targetome. Gene set enrichment analysis were used to integrate target genes in KEGG pathways and Protein-Protein Interaction networks (PPINs) and modules were built. Results. miR-26a-5p exerted an anti-proliferative effect acting at several levels, by decreasing survival and migration and inducing both cell cycle block and apoptosis. The analysis of the miR-26a-5p/targetome showed that 1423 (1352 coding and 71 non-coding) transcripts interacted with miR-26a-5p. Filtering the miR-26a-5p/targetome with prediction algorithms, 628 out of 1353 transcripts were miR-26a-5p predicted targets and 73 of them were already validated miR-26a-5p targets. Finally, miR-26a-5p targets were involved in 22 KEGG pathways and 20 significant protein-protein interaction modules Conclusion. The TS-miR-26a-5p/targetome is a platform that shows both unknown and known miRNA/target interactions thus offering the possibility to validate genes and discover pathways in which these genes could be involved.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA