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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cancer Genomics Proteomics ; 16(6): 433-442, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31659098

RESUMO

BACKGROUND/AIM: hERG potassium channels enhance tumor invasiveness and breast cancer proliferation. MicroRNA (miRNA) dysregulation during cancer controls gene regulation. The objective of this study was to identify miRNAs that regulate hERG expression in breast cancer. MATERIALS AND METHODS: Putative miRNAs targeting hERG were identified by bioinformatic approaches and screened using a 3'UTR luciferase assay. Functional assessments of endogenous hERG regulation were made using whole-cell electrophysiology, proliferation assays, and cell-cycle analyses following miRNA, hERG siRNA, or control transfection. RESULTS: miR-362-3p targeted hERG 3'UTR and was associated with higher survival rates in patients with breast cancer (HR=0.39, 95%CI=0.18-0.82). Enhanced miR-362-3p expression reduced hERG expression, peak current, and cell proliferation in cultured breast cancer cells (p<0.05). CONCLUSION: miR-362-3p mediates the transcriptional regulation of hERG and is associated with survival in breast cancer. The potential for miR-362-3p to serve as a biomarker and inform therapeutic strategies warrants further investigation.


Assuntos
Neoplasias da Mama/metabolismo , Proliferação de Células , Canal de Potássio ERG1/biossíntese , Potenciais da Membrana , MicroRNAs/metabolismo , Proteínas de Neoplasias/biossíntese , RNA Neoplásico/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Canal de Potássio ERG1/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , MicroRNAs/genética , Proteínas de Neoplasias/genética , RNA Neoplásico/genética
2.
Int J Cardiol ; 240: 367-371, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28433559

RESUMO

BACKGROUND: Long QT Syndrome type 2 (LQT2) is caused by mutations in the KCNH2 gene that encodes for the α-subunit (hERG) of the ion channel conducting the rapid delayed rectifier potassium current (IKr). We have previously identified a disease causing mutation (IVS9-28A/G) in the branch point of the splicing of KCNH2 intron 9. However, the mechanism through which this mutation causes the disease is unknown. METHODS AND RESULTS: We generated human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) from fibroblasts of two IVS9-28A/G mutation carriers. IVS9-28A/G iPSC-CMs showed prolonged repolarization time, mimicking what observed at the ECG level in the same patients. The expression of the full-length ERG1a isoform resulted reduced, whereas the C-terminally truncated ERG1aUSO isoform was upregulated in mutant iPSC-CMs, with consequent alteration of the physiological ERG1aUSO/ERG1a ratio. Importantly, we observed an impairment of hERG trafficking to the cell membrane. The severity of the alterations in hERG expression and trafficking correlated with the clinical severity of the disease in the two patients under study. Finally, we were able to revert the trafficking defect and reduce the repolarization duration in LQT2 iPSC-CMs using the proteasome inhibitor ALLN. CONCLUSION: Our results highlight the key role of the KCNH2 intron 9 branch point in the regulation of KCNH2 isoform expression and hERG channel function, and allow to categorize the IVS9-28A/G mutation as LQT2 class 2 mutation. These findings may result in a more personalized clinical management of IVS9-28A/G mutation carriers.


Assuntos
Canal de Potássio ERG1/biossíntese , Canal de Potássio ERG1/genética , Síndrome do QT Longo/genética , Síndrome do QT Longo/metabolismo , Mutação/genética , Miócitos Cardíacos/metabolismo , Feminino , Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Síndrome do QT Longo/patologia , Masculino , Miócitos Cardíacos/patologia , Linhagem , Transporte Proteico/fisiologia
3.
Int J Cancer ; 140(8): 1870-1880, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28120505

RESUMO

The role of progenitor/stem cells in pituitary tumorigenesis, resistance to pharmacological treatments and tumor recurrence is still unclear. This study investigated the presence of progenitor/stem cells in non-functioning pituitary tumors (NFPTs) and tested the efficacy of dopamine receptor type 2 (DRD2) and somatostatin receptor type 2 (SSTR2) agonists to inhibit in vitro proliferation. They found that 70% of 46 NFPTs formed spheres co-expressing stem cell markers, transcription factors (DAX1, SF1, ERG1) and gonadotropins. Analysis of tumor behavior showed that spheres formation was associated with tumor invasiveness (OR = 3,96; IC: 1.05-14.88, p = 0.036). The in vitro reduction of cell proliferation by DRD2 and SSTR2 agonists (31 ± 17% and 35 ± 13% inhibition, respectively, p < 0.01 vs. basal) occurring in about a half of NFPTs cells was conserved in the corresponding spheres. Accordingly, these drugs increased cyclin-dependent kinase inhibitor p27 and decreased cyclin D3 expression in spheres. In conclusion, they provided further evidence for the existence of cells with a progenitor/stem cells-like phenotype in the majority of NFPTs, particularly in those with invasive behavior, and demonstrated that the antiproliferative effects of dopaminergic and somatostatinergic drugs were maintained in progenitor/stem-like cells.


Assuntos
Carcinogênese/genética , Recidiva Local de Neoplasia/tratamento farmacológico , Neoplasias Hipofisárias/tratamento farmacológico , Receptores de Dopamina D2/genética , Receptores de Somatostatina/genética , Adulto , Carcinogênese/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ciclina D3/biossíntese , Inibidor de Quinase Dependente de Ciclina p27/biossíntese , Receptor Nuclear Órfão DAX-1/biossíntese , Dopaminérgicos/administração & dosagem , Resistencia a Medicamentos Antineoplásicos/genética , Canal de Potássio ERG1/biossíntese , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Gonadotropinas/biossíntese , Humanos , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Neoplasias Hipofisárias/genética , Neoplasias Hipofisárias/patologia , Fatores de Processamento de RNA/biossíntese , Receptores de Dopamina D2/agonistas , Receptores de Somatostatina/agonistas , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/patologia
4.
Hematology ; 21(10): 583-592, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27077769

RESUMO

OBJECTIVES: Human ether-a-go-go-related gene (HERG) K+ channels are shown to be aberrantly expressed in a variety of cancer cells where they play roles in contributing to cancer progression. Myelodysplastic syndromes (MDS) are a group of clinical heterogeneous disorders characterized by bone marrow failure and dysplasia of blood cells. However, the involvement of HERG K+ channels in MDS development is poorly understood. METHODS: The expression of HERG K+ channels in untreated MDS, acute myeloid leukemia (AML) patients and the control group was detected by flow cytometry. The roles of HERG K+ channels in regulation of SKM-1 cell proliferation, apoptosis, and cell cycle were determined by CCK-8 assay and flow cytometry, respectively. RESULTS: We found that expression of HERG K+ channels in MDS patients was significantly higher than controls and was lower than AML. Percentage of HERG K+ channels on CD34+CD38- cells gradually increased from controls to high-grade MDS subtypes. And HERG K+ channel levels showed an ascending tendency from low-risk to high-risk MDS group. In addition, the CCK-8 assay, apoptosis and cell cycle analysis were performed and showed that blockage of HERG K+ channels decreased the proliferation of MDS cells but rarely had effects on cell apoptosis and cell cycle distribution. CONCLUSION: Our study demonstrated that HERG K+ channels might be a potential tumor marker of MDS. These channels were likely to contribute to MDS progression and were helpful for predicting prognosis of MDS. Inhibition of HERG K+ channels might be a novel therapeutic measure for MDS.


Assuntos
Canal de Potássio ERG1/biossíntese , Síndromes Mielodisplásicas/metabolismo , Adolescente , Adulto , Idoso , Apoptose/fisiologia , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Ciclo Celular/fisiologia , Proliferação de Células/fisiologia , Criança , Canal de Potássio ERG1/genética , Feminino , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Masculino , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/patologia , Prognóstico , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA