Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Int J Mol Sci ; 19(3)2018 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-29543748

RESUMO

Glioblastomas (GBM) are the most frequent and aggressive brain tumors. In these malignancies, progesterone (P4) promotes proliferation, migration, and invasion. The P4 metabolite allopregnanolone (3α-THP) similarly promotes cell proliferation in the U87 human GBM cell line. Here, we evaluated global changes in gene expression of U87 cells treated with 3α-THP, P4, and the 5α-reductase inhibitor, finasteride (F). 3α-THP modified the expression of 137 genes, while F changed 90. Besides, both steroids regulated the expression of 69 genes. After performing an over-representation analysis of gene ontology terms, we selected 10 genes whose products are cytoskeleton components, transcription factors, and proteins involved in the maintenance of DNA stability and replication to validate their expression changes by RT-qPCR. 3α-THP up-regulated six genes, two of them were also up-regulated by F. Two genes were up-regulated by P4 alone, however, such an effect was blocked by F when cells were treated with both steroids. The remaining genes were regulated by the combined treatments of 3α-THP + F or P4 + F. An in-silico analysis revealed that promoters of the six up-regulated genes by 3α-THP possess cyclic adenosine monophosphate (cAMP) responsive elements along with CCAAT/Enhancer binding protein alpha (CEBPα) binding sites. These findings suggest that P4 and 3α-THP regulate different sets of genes that participate in the growth of GBMs.


Assuntos
Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Pregnanolona/farmacologia , Transcriptoma/efeitos dos fármacos , Inibidores de 5-alfa Redutase/farmacologia , Linhagem Celular Tumoral , Citoesqueleto/genética , Citoesqueleto/metabolismo , Finasterida/farmacologia , Humanos , Regiões Promotoras Genéticas , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Regulação para Cima
2.
Channels (Austin) ; 18(1): 2313323, 2024 12.
Artigo em Inglês | MEDLINE | ID: mdl-38354101

RESUMO

The members of the superfamily of Transient Receptor Potential (TRP) ion channels are physiologically important molecules that have been studied for many years and are still being intensively researched. Among the vanilloid TRP subfamily, the TRPV4 ion channel is an interesting protein due to its involvement in several essential physiological processes and in the development of various diseases. As in other proteins, changes in its function that lead to the development of pathological states, have been closely associated with modification of its regulation by different molecules, but also by the appearance of mutations which affect the structure and gating of the channel. In the last few years, some structures for the TRPV4 channel have been solved. Due to the importance of this protein in physiology, here we discuss the recent progress in determining the structure of the TRPV4 channel, which has been achieved in three species of animals (Xenopus tropicalis, Mus musculus, and Homo sapiens), highlighting conserved features as well as key differences among them and emphasizing the binding sites for some ligands that play crucial roles in its regulation.


Assuntos
Canais de Cátion TRPV , Canais de Potencial de Receptor Transitório , Camundongos , Animais , Canais de Cátion TRPV/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Mutação , Xenopus/metabolismo , Sítios de Ligação
3.
Function (Oxf) ; 5(5)2024 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-38984987

RESUMO

Polycystic kidney disease (PKD), a disease characterized by the enlargement of the kidney through cystic growth is the fourth leading cause of end-stage kidney disease world-wide. Transient receptor potential Vanilloid 4 (TRPV4), a calcium-permeable TRP, channel participates in kidney cell physiology and since TRPV4 forms complexes with another channel whose malfunction is associated to PKD, TRPP2 (or PKD2), we sought to determine whether patients with PKD, exhibit previously unknown mutations in TRPV4. Here, we report the presence of mutations in the TRPV4 gene in patients diagnosed with PKD and determine that they produce gain-of-function (GOF). Mutations in the sequence of the TRPV4 gene have been associated to a broad spectrum of neuropathies and skeletal dysplasias but not PKD, and their biophysical effects on channel function have not been elucidated. We identified and examined the functional behavior of a novel E6K mutant and of the previously known S94L and A217S mutant TRVP4 channels. The A217S mutation has been associated to mixed neuropathy and/or skeletal dysplasia phenotypes, however, the PKD carriers of these variants had not been diagnosed with these reported clinical manifestations. The presence of certain mutations in TRPV4 may influence the progression and severity of PKD through GOF mechanisms. PKD patients carrying TRVP4 mutations are putatively more likely to require dialysis or renal transplant as compared to those without these mutations.


Assuntos
Doenças Renais Policísticas , Canais de Cátion TRPV , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo , Humanos , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/patologia , Mutação , Feminino , Masculino , Células HEK293 , Mutação com Ganho de Função , Canais de Cátion TRPP/genética , Adulto
4.
Brain Sci ; 11(5)2021 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-33925221

RESUMO

Epithelial-mesenchymal transition (EMT) is an essential mechanism contributing to glioblastoma multiforme (GBM) progression, the most common and malignant brain tumor. EMT is induced by signaling pathways that crosstalk and regulate an intricate regulatory network of transcription factors. It has been shown that downstream components of 17ß-estradiol (E2) and transforming growth factor ß (TGF-ß) signaling pathways crosstalk in estrogen-sensitive tumors. However, little is known about the interaction between the E2 and TGF-ß signaling components in brain tumors. We have investigated the relationship between E2 and TGF-ß signaling pathways and their effects on EMT induction in human GBM-derived cells. Here, we showed that E2 and TGF-ß negatively regulated the expression of estrogen receptor α (ER-α) and Smad2/3. TGF-ß induced Smad2 phosphorylation and its subsequent nuclear translocation, which E2 inhibited. Both TGF-ß and E2 induced cellular processes related to EMT, such as morphological changes, actin filament reorganization, and mesenchymal markers (N-cadherin and vimentin) expression. Interestingly, we found that the co-treatment of E2 and TGF-ß blocked EMT activation. Our results suggest that E2 and TGF-ß signaling pathways interact through ER-α and Smad2/3 mediators in cells derived from human GBM and inhibit EMT activation induced by both factors alone.

5.
Methods Mol Biol ; 2174: 19-29, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32813242

RESUMO

Glioblastomas (GBM) are the most frequent and aggressive brain tumors due to their recurrence and resistance to current therapies. These characteristics are associated with the presence of glioma stem cells (GSCs), mainly identified by the detection of the membrane antigens CD133 and CD15. The main source of GSCs has been biopsies of tumors. However, alternatives are sought from cell lines because more homogeneous populations can be obtained with high yields. This chapter describes a method for the enrichment and characterization of GSCs from cell lines derived from human GBM by selective culture with serum-free neural stem cell medium and growth factors. The technique offers alternatives for the enrichment and characterization of GSCs, that could contribute to a better understanding of the biology of GBMs.


Assuntos
Neoplasias Encefálicas/patologia , Regulação Neoplásica da Expressão Gênica/genética , Glioblastoma/patologia , Células-Tronco Neoplásicas/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Antígeno AC133/análise , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Meios de Cultura Livres de Soro/química , Meios de Cultura Livres de Soro/farmacologia , Citometria de Fluxo , Glioblastoma/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Antígenos CD15/análise , Células-Tronco Neoplásicas/fisiologia , Células-Tronco Neurais/citologia
6.
Steroids ; 163: 108708, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32730775

RESUMO

Glioblastomas (GBMs) are the most common and deadliest intracranial tumors. Steroid hormones, such as progesterone (P4), at physiological concentrations, promote proliferation, and migration of human GBM cells in vivo and in vitro. Neuronal and glial cells, but also GBMs, metabolize P4 and synthesize different active metabolites such as 5α-dihydroprogesterone (5α-DHP). However, their contribution to GBM malignancy remains unknown. Here, we determined the 5α-DHP effects on the number of cells, proliferation, and migration of the U87 and U251 human GBM-derived cell lines. Of the tested concentrations (1 nM-1 µM), 5α-DHP 10 nM significantly increased the number of U87 and U251 cells from day 2 of treatment, and proliferation (at day 3) in a similar manner as P4 (10 nM). The treatment with the progesterone receptor (PR) antagonist RU486 (mifepristone), blocked the effects of 5α-DHP on the number of cells and proliferation. Besides, in U251 and LN229 GBM cells, 5α-DHP promoted cell migration (from 12 to 24 h). We also determined that GBM cells expressed the 3α-hydroxysteroid oxidoreductases (3α-HSOR), which reversibly reduce 5α-DHP to allopregnanolone (3α-THP). These data indicate that 5α-DHP induces proliferation and migration of human GBM through the activation of PR.


Assuntos
5-alfa-Di-Hidroprogesterona/farmacologia , Antineoplásicos/farmacologia , Movimento Celular/efeitos dos fármacos , Glioblastoma/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Receptores de Progesterona/metabolismo
7.
Cells ; 9(9)2020 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-32825553

RESUMO

The mesenchymal phenotype of glioblastoma multiforme (GBM), the most frequent and malignant brain tumor, is associated with the worst prognosis. The epithelial-mesenchymal transition (EMT) is a cell plasticity mechanism involved in GBM malignancy. In this study, we determined 17ß-estradiol (E2)-induced EMT by changes in cell morphology, expression of EMT markers, and cell migration and invasion assays in human GBM-derived cell lines. E2 (10 nM) modified the shape and size of GBM cells due to a reorganization of actin filaments. We evaluated EMT markers expression by RT-qPCR, Western blot, and immunofluorescence.We found that E2 upregulated the expression of the mesenchymal markers, vimentin, and N-cadherin. Scratch and transwell assays showed that E2 increased migration and invasion of GBM cells. The estrogen receptor-α (ER-α)-selective agonist 4,4',4''-(4-propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol (PPT, 10 nM) affected similarly to E2 in terms of the expression of EMT markers and cell migration, and the treatment with the ER-α antagonist methyl-piperidino-pyrazole (MPP, 1 µM) blocked E2 and PPT effects. ER-ß-selective agonist diarylpropionitrile (DNP, 10 nM) and antagonist 4-[2-phenyl-5,7-bis(trifluoromethyl)pyrazole[1,5-a]pyrimidin-3-yl]phenol (PHTPP, 1 µM) showed no effects on EMT marker expression. These data suggest that E2 induces EMT activation through ER-α in human GBM-derived cells.


Assuntos
Transição Epitelial-Mesenquimal/efeitos dos fármacos , Estradiol/uso terapêutico , Estrogênios/uso terapêutico , Glioblastoma/tratamento farmacológico , Estradiol/farmacologia , Estrogênios/farmacologia , Glioblastoma/patologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA