Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Toxicol Appl Pharmacol ; 305: 12-21, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27245768

RESUMO

The effects of some progestins on fish reproduction have been recently reported revealing the hazard of this class of steroidal pharmaceuticals. However, their effects at the central nervous system level have been poorly studied until now. Notwithstanding, progesterone, although still widely considered primarily a sex hormone, is an important agent affecting many central nervous system functions. Herein, we investigated the effects of a large set of synthetic ligands of the nuclear progesterone receptor on the glial-specific expression of the zebrafish brain aromatase (cyp19a1b) using zebrafish mechanism-based assays. Progesterone and 24 progestins were first screened on transgenic cyp19a1b-GFP zebrafish embryos. We showed that progesterone, dydrogesterone, drospirenone and all the progesterone-derived progestins had no effect on GFP expression. Conversely, all progestins derived from 19-nortesterone induced GFP in a concentration-dependent manner with EC50 ranging from the low nM range to hundreds nM. The 19-nortestosterone derived progestins levonorgestrel (LNG) and norethindrone (NET) were further tested in a radial glial cell context using U251-MG cells co-transfected with zebrafish ER subtypes (zfERα, zfERß1 or zfERß2) and cyp19a1b promoter linked to luciferase. Progesterone had no effect on luciferase activity while NET and LNG induced luciferase activity that was blocked by ICI 182,780. Zebrafish-ERs competition assays showed that NET and LNG were unable to bind to ERs, suggesting that the effects of these compounds on cyp19a1b require metabolic activation prior to elicit estrogenic activity. Overall, we demonstrate that 19-nortestosterone derived progestins elicit estrogenic activity by inducing cyp19a1b expression in radial glial cells. Given the crucial role of radial glial cells and neuro-estrogens in early development of brain, the consequences of exposure of fish to these compounds require further investigation.


Assuntos
Aromatase/metabolismo , Neuroglia/efeitos dos fármacos , Congêneres da Progesterona/farmacologia , Proteínas de Peixe-Zebra/metabolismo , Androgênios/farmacologia , Animais , Animais Geneticamente Modificados , Aromatase/genética , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Linhagem Celular Tumoral , Estradiol/farmacologia , Estrogênios/farmacologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Neuroglia/metabolismo , Receptores de Estrogênio/metabolismo , Testosterona/farmacologia , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
2.
Biochem J ; 461(2): 257-68, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24762104

RESUMO

In addition to soluble factors, mechanical constraints and extracellular matrix stiffness are important regulators of cell fate that are mediated by cytoskeletal modifications. The EMT (epithelial-mesenchymal transition) that occurs during normal development and malignant progression is a typical example of the phenotypic switch associated with profound actin remodelling and changes in gene expression. For instance, actin dynamics control motile cell functions in EMT, in part, through regulating the subcellular localization of the myocardin-related transcription factor MKL1 (megakaryoblastic leukaemia translocation 1), a co-activator of SRF (serum-responsive factor). In the present paper, we show that MKL1 participates also to the control of the cellular switch between growth and quiescence. Experimental disconnection between MKL1 and G-actin (globular actin), by using an MKL1 mutant or enhancing the F (filamentous)-/G-actin ratio, generates a widely open chromatin state and a global increase in biosynthetic activity, classically associated with cell growth. Conversely, G-actin accumulation favours nuclear condensation and cell quiescence. These large-scale chromatin changes rely upon extensive histone modifications, exemplified by that of H3K9 (H3 Lys9) shifting from trimethylation, a heterochromatin mark, to acetylation, a mark of euchromatin. The present study provides the first evidence for a global reversible hetero/euchromatinization phenomenon triggered by the actin/MKL1 signalling pathway.


Assuntos
Actinas/metabolismo , Montagem e Desmontagem da Cromatina , Proteínas de Ligação a DNA/metabolismo , Histonas/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Processamento de Proteína Pós-Traducional , Acetilação , Actinas/genética , Linhagem Celular Tumoral , Proliferação de Células , Cromatina/química , Proteínas de Ligação a DNA/genética , Transição Epitelial-Mesenquimal/genética , Expressão Gênica , Histonas/genética , Humanos , Metilação , Proteínas de Fusão Oncogênica/genética , Transdução de Sinais , Transativadores
3.
Mol Cell Endocrinol ; 530: 111282, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-33894309

RESUMO

The Myocardin-related transcription factor A [MRTFA, also known as Megakaryoblastic Leukemia 1 (MKL1))] is a major actor in the epithelial to mesenchymal transition (EMT). We have previously shown that activation and nuclear accumulation of MRTFA mediate endocrine resistance of estrogen receptor alpha (ERα) positive breast cancers by initiating a partial transition from luminal to basal-like phenotype and impairing ERα cistrome and transcriptome. In the present study, we deepen our understanding of the mechanism by monitoring functional changes in the receptor's activity. We demonstrate that MRTFA nuclear accumulation down-regulates the expression of the unliganded (Apo-)ERα and causes a redistribution of the protein localization from its normal nuclear place to the entire cell volume. This phenomenon is accompanied by a shift in Apo-ERα monomer/dimer ratio towards the monomeric state, leading to significant functional consequences on ERα activities. In particular, the association of Apo-ERα with chromatin is drastically decreased, and the remaining ERα binding sites are substantially less enriched in ERE motifs than in control conditions. Monitored by proximity Ligation Assay, ERα interactions with P160 family coactivators are partly impacted when MRTFA accumulates in the nucleus, and those with SMRT and NCOR1 corepressors are abolished. Finally, ERα interactions with kinases such as c-src and PI3K are increased, thereby enhancing MAP Kinase and AKT activities. In conclusion, the activation and nuclear accumulation of MRTFA in ERα positive breast cancer cells remodels both ERα location and functions by shifting its activity from nuclear genome regulation to extra-nuclear non-genomic signaling.


Assuntos
Neoplasias da Mama/metabolismo , Núcleo Celular/metabolismo , Receptor alfa de Estrogênio/metabolismo , Transativadores/genética , Transativadores/metabolismo , Sítios de Ligação , Neoplasias da Mama/genética , Cromatina/metabolismo , Transição Epitelial-Mesenquimal , Receptor alfa de Estrogênio/química , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Transporte Proteico
4.
J Biol Chem ; 284(49): 33729-39, 2009 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-19826002

RESUMO

Although involved in processes leading to the emergence and development of hormone-dependent breast cancers, the estrogen receptor alpha (ERalpha) also prevents transformed cells from progressing toward a more aggressive phenotype. The transcriptional activity of ERalpha is mediated through two transactivation functions, called activation function 1 and 2, whose respective involvement varies in a cell-specific manner. Here, we identify the Rho/megakaryoblastic leukemia 1 (MKL1) signaling pathway as a main actor in controlling the cell-specific activity of both transactivation functions of ERalpha. Notably, we show that, when the coregulator MKL1 is sequestered in an inactive form by unpolymerized actin, the transcriptional activity of ERalpha mainly relies on the activation function 1. The activation of MKL1, which results from its dissociation from unpolymerized actin, promoted by the ability of Rho to support polymeric actin accumulation, silences the activation function 1 of ERalpha and allows the receptor to mainly act through its activation function 2. Importantly, this switch in the respective contribution exerted by both transactivation functions is correlated with an impaired ability of ERalpha to efficiently transactivate estrogen-regulated reporter genes. MKL1 is further shown to be present on estrogen-responsive genes in vivo. Interestingly, the Rho/MKL1 signaling pathway is activated during the epithelial-mesenchymal transition. A reduced transactivation efficiency of ERalpha, resulting from the activation of this pathway, may therefore suppress the protective role exerted by ERalpha toward tumor progression and invasiveness.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Receptor alfa de Estrogênio/metabolismo , Regulação da Expressão Gênica , Leucemia Megacarioblástica Aguda/metabolismo , Proteínas de Fusão Oncogênica/biossíntese , Proteínas rho de Ligação ao GTP/biossíntese , Actinas/química , Linhagem Celular Tumoral , DNA/química , Proteínas de Ligação a DNA/metabolismo , Células HeLa , Humanos , Proteínas de Fusão Oncogênica/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Transdução de Sinais , Transativadores , Transcrição Gênica , Ativação Transcricional , Proteínas rho de Ligação ao GTP/metabolismo
5.
Biochem Biophys Res Commun ; 365(2): 304-9, 2008 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-17991426

RESUMO

The estrogen receptor alpha (ER alpha) is key in regulating normal breast development and function and is closely involved in the onset and progress of cancers. ER alpha transcriptional activity is mediated through two activation functions, AF1 and AF2, whose activity is tightly regulated in a cell-specific manner through yet unknown processes. Here, we demonstrate that cell-cell junctions generate cell permissiveness to AF1 through an up-regulation of the activity of an AF1 sub-region termed box 1. Moreover, the loss of E-cadherin expression is shown to silence the AF1 activity of ER alpha, allowing the receptor to mainly act through its AF2. This switch from an AF1 to an AF2 cell permissiveness also consequently results in the attenuation of ER alpha activity. Therefore, a loss of cell-cell junctions, a key process that occurs during the epithelial-mesenchymal transition, should have a broad impact on ER alpha transcriptional functions.


Assuntos
Caderinas/metabolismo , Receptor alfa de Estrogênio/metabolismo , Furilfuramida/metabolismo , Hepatócitos/metabolismo , Junções Intercelulares/fisiologia , Fatores de Transcrição/metabolismo , Ativação Transcricional/fisiologia , Linhagem Celular , Células HeLa , Humanos
6.
Endocrinology ; 146(12): 5474-84, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16150902

RESUMO

The expression of two human estrogen receptor-alpha (hERalpha) isoforms has been characterized within estrogen receptor-alpha-positive breast cancer cell lines such as MCF7: the full-length hERalpha66 and the N terminally deleted hERalpha46, which is devoid of activation function (AF)-1. Although hERalpha66 is known to mediate the mitogenic effects that estrogens have on MCF7 cells, the exact function of hERalpha46 in these cells remains undefined. Here we show that, during MCF7 cell growth, hERalpha46 is mainly expressed in the nucleus at relatively low levels, whereas hERalpha66 accumulates in the nucleus. When cells reach confluence, the situation reverses, with hERalpha46 accumulating within the nucleus. Although hERalpha46 expression remains rather stable during an estrogen-induced cell cycle, its overexpression in proliferating MCF7 cells provokes a cell-cycle arrest in G(0)/G(1) phases. To gain further details on the influence of hERalpha46 on cell growth, we used PC12 estrogen receptor-alpha-negative cell line, in which stable transfection of hERalpha66 but not hERalpha46 allows estrogens to behave as mitogens. We next demonstrate that, in MCF7 cells, overexpression of hERalpha46 inhibits the hERalpha66-mediated estrogenic induction of all AF-1-sensitive reporters: c-fos and cyclin D1 as well as estrogen-responsive element-driven reporters. Our data indicate that this inhibition occurs likely through functional competitions between both isoforms. In summary, hERalpha46 antagonizes the proliferative action of hERalpha66 in MCF7 cells in part by inhibiting hERalpha66 AF-1 activity.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Receptor alfa de Estrogênio/antagonistas & inibidores , Receptor alfa de Estrogênio/metabolismo , Ligação Competitiva , Neoplasias da Mama/genética , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proliferação de Células , Dimerização , Estradiol/farmacologia , Estrogênios , Feminino , Fase G1 , Regulação da Expressão Gênica , Humanos , Elementos de Resposta , Fase de Repouso do Ciclo Celular , Distribuição Tecidual
7.
Endocrinology ; 143(7): 2571-83, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12072389

RESUMO

In this study, we hypothesized that many of the reported effects of phthalate esters and other peroxisome proliferators (PPs) in the testis are mediated by members of the PP- activated receptor (PPAR) family of transcription factors through alterations in proteins involved in steroidogenesis. Exposure of Leydig cells to PPs prevented cholesterol transport into the mitochondria after hormonal stimulation and inhibited steroid synthesis, without altering total cell protein synthesis or mitochondrial and DNA integrity. PPs also reduced the levels of the cholesterol-binding protein peripheral-type benzodiazepine receptor (PBR) because of a direct transcriptional inhibition of PBR gene expression in MA-10 Leydig cells. MA-10 cells contain mRNAs for PPARalpha and PPARbeta/delta, but not for PPARgamma. In vivo treatment of mice with PPs resulted in the reduction of both testis PBR mRNA and circulating testosterone levels, in agreement with the proposed role of PBR in steroidogenesis. By contrast, liver PBR mRNA levels were increased, in agreement with the proposed role of PBR in cell growth/tumor formation in nonsteroidogenic tissues. However, PPs did not inhibit testosterone production and testis PBR expression in PPARalpha-null mice. These results suggest that the antiandrogenic effect of PPs is mediated by a PPARalpha-dependent inhibition of Leydig cell PBR gene expression.


Assuntos
Antagonistas de Androgênios , Colesterol/metabolismo , Células Intersticiais do Testículo/metabolismo , Proliferadores de Peroxissomos/farmacologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de GABA-A/biossíntese , Esteroides/biossíntese , Fatores de Transcrição/metabolismo , Animais , Transporte Biológico Ativo/efeitos dos fármacos , Northern Blotting , Proteínas de Transporte/biossíntese , Proteínas de Transporte/genética , Catalase/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Gonadotropina Coriônica/antagonistas & inibidores , Dano ao DNA/efeitos dos fármacos , Eletroforese em Gel de Poliacrilamida , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Células Intersticiais do Testículo/efeitos dos fármacos , Masculino , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Elastase Pancreática , RNA Mensageiro/biossíntese , Radioimunoensaio , Ensaio Radioligante , Ratos , Receptores Citoplasmáticos e Nucleares/efeitos dos fármacos , Receptores de GABA-A/efeitos dos fármacos , Receptores de GABA-A/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/efeitos dos fármacos , Transfecção
8.
J Mol Med (Berl) ; 92(11): 1179-200, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25081415

RESUMO

UNLABELLED: The existence of a link between estrogen deprivation and osteoarthritis (OA) in postmenopausal women suggests that 17ß-estradiol (17ß-E2) may be a modulator of cartilage homeostasis. Here, we demonstrate that 17ß-E2 stimulates, via its receptor human estrogen receptor α 66 (hERα66), type II collagen expression in differentiated and dedifferentiated (reflecting the OA phenotype) articular chondrocytes. Transactivation of type II collagen gene (COL2A1) by ligand-independent transactivation domain (AF-1) of hERα66 was mediated by "GC" binding sites of the -266/-63-bp promoter, through physical interactions between ERα, Sp1/Sp3, Sox9, and p300, as demonstrated in chromatin immunoprecipitation (ChIP) and Re-Chromatin Immuno-Precipitation (Re-ChIP) assays in primary and dedifferentiated cells. 17ß-E2 and hERα66 increased the DNA-binding activities of Sp1/Sp3 and Sox-9 to both COL2A1 promoter and enhancer regions. Besides, Sp1, Sp3, and Sox-9 small interfering RNAs (siRNAs) prevented hERα66-induced transactivation of COL2A1, suggesting that these factors and their respective cis-regions are required for hERα66-mediated COL2A1 up-regulation. Our results highlight the genomic pathway by which 17ß-E2 and hERα66 modulate Sp1/Sp3 heteromer binding activity and simultaneously participate in the recruitment of the essential factors Sox-9 and p300 involved respectively in the chondrocyte-differentiated status and COL2A1 transcriptional activation. These novel findings could therefore be attractive for tissue engineering of cartilage in OA, by the fact that 17ß-E2 could promote chondrocyte redifferentiation. KEY MESSAGES: 17ß-E2 up-regulates type II collagen gene expression in articular chondrocytes. An ERα66/Sp1/Sp3/Sox-9/p300 protein complex mediates this stimulatory effect. This heteromeric complex interacts and binds to Col2a1 promoter and enhancer in vivo. Our findings highlight a new regulatory mechanism for 17ß-E2 action in chondrocytes. 17ß-E2 might be an attractive candidate for cartilage engineering applications.


Assuntos
Condrócitos/citologia , Colágeno Tipo II/metabolismo , Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Fatores de Transcrição SOX9/metabolismo , Fator de Transcrição Sp1/metabolismo , Fator de Transcrição Sp3/metabolismo , Animais , Sítios de Ligação , Cartilagem Articular/citologia , Diferenciação Celular , Colágeno Tipo II/genética , Humanos , Masculino , Fenótipo , Regiões Promotoras Genéticas , RNA Interferente Pequeno/metabolismo , Coelhos , Ativação Transcricional , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA