Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 337
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 49(22): 12716-12731, 2021 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-34850111

RESUMO

Here, we report that in T47D breast cancer cells 50 pM progestin is sufficient to activate cell cycle entry and the progesterone gene expression program. At this concentration, equivalent to the progesterone blood levels found around the menopause, progesterone receptor (PR) binds only to 2800 genomic sites, which are accessible to ATAC cleavage prior to hormone exposure. These highly accessible sites (HAs) are surrounded by well-organized nucleosomes and exhibit breast enhancer features, including estrogen receptor alpha (ERα), higher FOXA1 and BRD4 (bromodomain containing 4) occupancy. Although HAs are enriched in RAD21 and CTCF, PR binding is the driving force for the most robust interactions with hormone-regulated genes. HAs show higher frequency of 3D contacts among themselves than with other PR binding sites, indicating colocalization in similar compartments. Gene regulation via HAs is independent of classical coregulators and ATP-activated remodelers, relying mainly on MAP kinase activation that enables PR nuclear engagement. HAs are also preferentially occupied by PR and ERα in breast cancer xenografts derived from MCF-7 cells as well as from patients, indicating their potential usefulness as targets for therapeutic intervention.


Assuntos
Neoplasias da Mama/genética , Elementos Facilitadores Genéticos , Regulação Neoplásica da Expressão Gênica , Progestinas/fisiologia , Animais , Neoplasias da Mama/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células , Cromatina , Receptor alfa de Estrogênio/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases , Células MCF-7 , Camundongos , Promegestona/farmacologia , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo
2.
Nucleic Acids Res ; 47(20): 10645-10661, 2019 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-31598691

RESUMO

The glucocorticoid and progesterone receptors (GR and PR) are closely related members of the steroid receptor family. Despite sharing similar structural and functional characteristics; the cognate hormones display very distinct physiological responses. In mammary epithelial cells, PR activation is associated with the incidence and progression of breast cancer, whereas the GR is related to growth suppression and differentiation. Despite their pharmacological relevance, only a few studies have compared GR and PR activities in the same system. Using a PR+/GR+ breast cancer cell line, here we report that either glucocorticoid-free or dexamethasone (DEX)-activated GR inhibits progestin-dependent gene expression associated to epithelial-mesenchymal-transition and cell proliferation. When both receptors are activated with their cognate hormones, PR and GR can form part of the same complex according to co-immunoprecipitation, quantitative microscopy and sequential ChIP experiments. Moreover, genome-wide studies in cells treated with either DEX or R5020, revealed the presence of several regions co-bound by both receptors. Surprisingly, GR also binds novel genomic sites in cells treated with R5020 alone. This progestin-induced GR binding was enriched in REL DNA motifs and located close to genes coding for chromatin remodelers. Understanding GR behavior in the context of progestin-dependent breast cancer could provide new targets for tumor therapy.


Assuntos
Neoplasias da Mama/genética , Regulação Neoplásica da Expressão Gênica , Genoma Humano , Receptores de Glucocorticoides/metabolismo , Receptores de Progesterona/metabolismo , Sequência de Bases , Sítios de Ligação , Neoplasias da Mama/patologia , Desdiferenciação Celular/efeitos dos fármacos , Desdiferenciação Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Cromatina/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/farmacologia , Humanos , Progestinas/farmacologia , Promegestona/farmacologia , Ligação Proteica/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos
3.
Eur J Contracept Reprod Health Care ; 26(3): 184-194, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33645377

RESUMO

PURPOSE: To evaluate the pharmacokinetics and pharmacodynamics of oestriol (E3) and trimegestone (TMG) in healthy women after application of three different vaginal rings over 21 days. The vaginal rings had a nominal delivery rate of 0.413/0.050 mg/day (Test 1), 0.311/0.090 mg/day (Test 2) and 0.209/0.137 mg/day (Test 3) E3/TMG. METHODS: Thirty-five healthy women were randomised to receive a single application of Test 1, 2 or 3 (Clinical Trial NCT03343912). The E3 and TMG plasma concentration was determined by LC-MS/MS. Oestradiol (E2) and progesterone (PG) serum concentrations, and bleeding patern were determined as pharmacodynamic parameters. Safety was assessed by evaluation of adverse events and local tolerability. RESULTS: The total and maximum exposure of E3 and TMG increased in a proportional ratio to dose. However, not in a magnitude which was expected from the dose differences for E3. During Test 2 and 3 treatment all E2 and PG values remained on a well suppressed level until end of treatment. E2 and PG serum levels increased distinctly earlier after ring removal with Test 1 compared to Test 2 and 3. Test 3 achieved 95.24% of "no bleeding" days under treatment followed by Test 1 (91.67%), and Test 2 (86.15%). CONCLUSIONS: The Test 3 formulation presented the best dose combination of E3/TMG for contraception. Moreover, all vaginal rings were well tolerated.


Assuntos
Anticoncepcionais Femininos/administração & dosagem , Dispositivos Anticoncepcionais Femininos , Estriol/farmacologia , Estriol/farmacocinética , Estrogênios/metabolismo , Promegestona/análogos & derivados , Administração Intravaginal , Adulto , Cromatografia Líquida , Estradiol/sangue , Estrogênios/sangue , Feminino , Humanos , Progesterona/sangue , Promegestona/farmacocinética , Promegestona/farmacologia , Espectrometria de Massas em Tandem
4.
Exp Cell Res ; 382(1): 111433, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31100306

RESUMO

Autophagy is an evolutionary conserved, self-eating process that targets cellular constituents for lysosomal degradation. Transcription factor EB (TFEB) is a master regulator of autophagy by inducing the expression of genes involved in autophagic and lysosomal degradation. In breast cancer, ligand-activated progesterone receptor has been reported to influence cancer development by manipulating the autophagy pathway. However, understanding of the mechanism that underlies this autophagic response remains limited. Herein, we report that prolonged treatment with progestin R5020 upregulates autophagy in MCF-7 human breast cancer cells via a novel interplay between progesterone receptor B (PRB) and TFEB. R5020 upregulates TFEB gene expression and protein levels in a PRB-dependent manner. Additionally, R5020 enhances the co-recruitment of PRB and TFEB to each other to facilitate TFEB nuclear localization. Once in the nucleus, TFEB induces the expression of autophagy and lysosomal genes to potentiate autophagy. Together, our findings highlight a novel functional connection between ligand-activated PRB and TFEB to modulate autophagy in MCF-7 breast cancer cells. As breast cancer development is controlled by autophagy, the progestin-PRB-TFEB transduction pathway warrants future attention as a potential therapeutic target in cancer therapy.


Assuntos
Adenocarcinoma/genética , Autofagia/fisiologia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/fisiologia , Neoplasias da Mama/genética , Proteínas de Neoplasias/fisiologia , Receptores de Progesterona/fisiologia , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Autofagossomos/metabolismo , Autofagia/genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/biossíntese , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Lisossomos/metabolismo , Células MCF-7 , Proteínas Associadas aos Microtúbulos/biossíntese , Proteínas Associadas aos Microtúbulos/genética , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Promegestona/farmacologia , Mapeamento de Interação de Proteínas , Transporte Proteico/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ativação Transcricional
5.
Genes Dev ; 25(8): 845-62, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21447625

RESUMO

Gene regulation by external signals requires access of transcription factors to DNA sequences of target genes, which is limited by the compaction of DNA in chromatin. Although we have gained insight into how core histones and their modifications influence this process, the role of linker histones remains unclear. Here we show that, within the first minute of progesterone action, a complex cooperation between different enzymes acting on chromatin mediates histone H1 displacement as a requisite for gene induction and cell proliferation. First, activated progesterone receptor (PR) recruits the chromatin remodeling complexes NURF and ASCOM (ASC-2 [activating signal cointegrator-2] complex) to hormone target genes. The trimethylation of histone H3 at Lys 4 by the MLL2/MLL3 subunits of ASCOM, enhanced by the hormone-induced displacement of the H3K4 demethylase KDM5B, stabilizes NURF binding. NURF facilitates the PR-mediated recruitment of Cdk2/CyclinA, which is required for histone H1 displacement. Cooperation of ATP-dependent remodeling, histone methylation, and kinase activation, followed by H1 displacement, is a prerequisite for the subsequent displacement of histone H2A/H2B catalyzed by PCAF and BAF. Chromatin immunoprecipitation (ChIP) and sequencing (ChIP-seq) and expression arrays show that H1 displacement is required for hormone induction of most hormone target genes, some of which are involved in cell proliferation.


Assuntos
Histona Desmetilases com o Domínio Jumonji/metabolismo , Proteínas Nucleares/metabolismo , Promegestona/farmacologia , Proteínas Repressoras/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo , Antígenos Nucleares/genética , Antígenos Nucleares/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Montagem e Desmontagem da Cromatina , Imunoprecipitação da Cromatina , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Histonas , Humanos , Imunoprecipitação , Histona Desmetilases com o Domínio Jumonji/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Interferência de RNA , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Proteínas Repressoras/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição de p300-CBP/genética
6.
BMC Cancer ; 16: 326, 2016 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-27215412

RESUMO

BACKGROUND: The synthesis of specific, potent progesterone antagonists adds potential agents to the breast cancer prevention and treatment armamentarium. The identification of individuals who will benefit from these agents will be a critical factor for their clinical success. METHODS: We utilized telapristone acetate (TPA; CDB-4124) to understand the effects of progesterone receptor (PR) blockade on proliferation, apoptosis, promoter binding, cell cycle progression, and gene expression. We then identified a set of genes that overlap with human breast luteal-phase expressed genes and signify progesterone activity in both normal breast cells and breast cancer cell lines. RESULTS: TPA administration to T47D cells results in a 30 % decrease in cell number at 24 h, which is maintained over 72 h only in the presence of estradiol. Blockade of progesterone signaling by TPA for 24 h results in fewer cells in G2/M, attributable to decreased expression of genes that facilitate the G2/M transition. Gene expression data suggest that TPA affects several mechanisms that progesterone utilizes to control gene expression, including specific post-translational modifications, and nucleosomal organization and higher order chromatin structure, which regulate access of PR to its DNA binding sites. CONCLUSIONS: By comparing genes induced by the progestin R5020 in T47D cells with those increased in the luteal-phase normal breast, we have identified a set of genes that predict functional progesterone signaling in tissue. These data will facilitate an understanding of the ways in which drugs such as TPA may be utilized for the prevention, and possibly the therapy, of human breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Redes Reguladoras de Genes/efeitos dos fármacos , Norpregnadienos/farmacologia , Promegestona/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Feminino , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células MCF-7 , Análise de Sequência com Séries de Oligonucleotídeos/métodos
7.
J Cell Physiol ; 230(7): 1594-606, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25536295

RESUMO

We previously encountered regulatory processes wherein dihydrotestosterone (DHT) exerted its inhibitory effect on parathyroid hormone-related protein (PTHrP) gene repression through the estrogen receptor (ER)α, but not the androgen receptor (AR), in breast cancer MCF-7 cells. Here, we investigated whether such aberrant ligand-nuclear receptor (NR) interaction is present in prostate cancer LNCaP cells. First, we confirmed that LNCaP cells expressed large amounts of AR at negligible levels of ERα/ß or progesterone receptor. Both suppression of PTHrP and activation of prostate-specific antigen genes were observed after independent administration of 17ß-estradiol (E2), DHT, or R5020. Consistent with the notion that the LNCaP AR lost its ligand specificity due to a mutation (Thr-Ala877), experiments with siRNA targeting the respective NR revealed that the AR monopolized the role of the mediator of shared hormone-dependent regulation, which was invariably associated with nuclear translocation of this mutant AR. Microarray analysis of gene regulation by DHT, E2, or R5020 disclosed that more than half of the genes downstream of the AR (Thr-Ala877) overlapped in the LNCaP cells. Of particular interest, we realized that the AR (wild-type [wt]) and AR (Thr-Ala877) were equally responsible for the E2-AR interactions. Fluorescence microscopy experiments demonstrated that both EGFP-AR (wt) and EGFP-AR (Thr-Ala877) were exclusively localized within the nucleus after E2 or DHT treatment. Furthermore, reporter assays revealed that some other cancer cells exhibited aberrant E2-AR (wt) signaling similar to that in the LNCaP cells. We herein postulate the presence of entangled interactions between wt AR and E2 in certain hormone-sensitive cancer cells.


Assuntos
Neoplasias da Mama/metabolismo , Estradiol/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Linhagem Celular Tumoral , Di-Hidrotestosterona/farmacologia , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Mutação , Proteína Relacionada ao Hormônio Paratireóideo/genética , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Promegestona/farmacologia , Receptores Androgênicos/genética
8.
Nucleic Acids Res ; 41(12): 6072-86, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23640331

RESUMO

Steroid receptors were classically described for regulating transcription by binding to target gene promoters. However, genome-wide studies reveal that steroid receptors-binding sites are mainly located at intragenic regions. To determine the role of these sites, we examined the effect of progestins on the transcription of the bcl-x gene, where only intragenic progesterone receptor-binding sites (PRbs) were identified. We found that in response to hormone treatment, the PR is recruited to these sites along with two histone acetyltransferases CREB-binding protein (CBP) and GCN5, leading to an increase in histone H3 and H4 acetylation and to the binding of the SWI/SNF complex. Concomitant, a more relaxed chromatin was detected along bcl-x gene mainly in the regions surrounding the intragenic PRbs. PR also mediated the recruitment of the positive elongation factor pTEFb, favoring RNA polymerase II (Pol II) elongation activity. Together these events promoted the re-distribution of the active Pol II toward the 3'-end of the gene and a decrease in the ratio between proximal and distal transcription. These results suggest a novel mechanism by which PR regulates gene expression by facilitating the proper passage of the polymerase along hormone-dependent genes.


Assuntos
RNA Polimerase II/metabolismo , Receptores de Progesterona/metabolismo , Elongação da Transcrição Genética , Proteína bcl-X/genética , Processamento Alternativo , Sítios de Ligação , Proteína de Ligação a CREB/metabolismo , Linhagem Celular Tumoral , Cromatina/química , Humanos , Fator B de Elongação Transcricional Positiva/metabolismo , Promegestona/farmacologia , Proteína bcl-X/biossíntese , Proteína bcl-X/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo
9.
Mol Hum Reprod ; 20(3): 222-34, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24219889

RESUMO

Previous microarray analyses indicated that a portion of the transcriptome in the macaque corpus luteum (CL) of the menstrual cycle was regulated indirectly by luteinizing hormone via the local actions of steroid hormones, notably progesterone (P). The current study was designed to investigate this concept in the CL of early pregnancy by analyzing chorionic gonadotrophin (CG)-regulated genes that are dependent versus independent of local steroid action. Exogenous human chorionic gonadotropin treatment simulating early pregnancy (SEP) began on Day 9 of the luteal phase in female rhesus monkeys with and without concurrent administration of the 3-ß-hydroxysteroid dehydrogenase inhibitor trilostane (TRL) with or without the synthetic progestin R5020. Compared with SEP treatment alone, TRL altered 50 mRNA transcripts on Day 10, rising to 95 on Day 15 (P<0.05, ≥2-fold change in gene expression). Steroid-sensitive genes were validated; notably effects of steroid ablation and P replacement varied by day. Expression of some genes previously identified as P-regulated in the macaque CL during the menstrual cycle were not significantly altered by steroid ablation and P replacement during CG exposure in SEP. These data indicate that the majority of CG-regulated luteal transcripts are differentially expressed independently of local steroid actions. However, the steroid-regulated genes in the macaque CL may be essential during early pregnancy, based on previous reports that TRL treatment initiates premature structural regression of the CL during SEP. These data reinforce the concept that the structure, function and regulation of the rescued CL in early pregnancy differs from the CL of the menstrual cycle in primates.


Assuntos
Abortivos Esteroides/farmacologia , Corpo Lúteo/efeitos dos fármacos , Di-Hidrotestosterona/análogos & derivados , Congêneres da Progesterona/farmacologia , Promegestona/farmacologia , RNA Mensageiro/genética , Transcriptoma/efeitos dos fármacos , 3-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , 3-Hidroxiesteroide Desidrogenases/genética , 3-Hidroxiesteroide Desidrogenases/metabolismo , Animais , Gonadotropina Coriônica/farmacologia , Corpo Lúteo/metabolismo , Di-Hidrotestosterona/farmacologia , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Fase Luteal/efeitos dos fármacos , Fase Luteal/genética , Hormônio Luteinizante/farmacologia , Macaca mulatta , Gravidez , RNA Mensageiro/metabolismo
10.
Reprod Biol Endocrinol ; 11: 33, 2013 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-23651709

RESUMO

BACKGROUND: The transport of gametes as well as the zygote is facilitated by motile cilia lining the inside of the fallopian tube. Progesterone reduces the ciliary beat frequency within 30 minutes in both cows and mice. This rapid reduction suggest the involvement of a non-genomic signaling mechanism, although it is not known which receptors that are involved. Here we investigated the possible involvement of the classical progesterone receptor in this process. METHOD: The ciliary beat frequency of mice fallopian tube was measured ex vivo using an inverted bright field microscope and a high speed camera. The effects of the agonists progesterone and promegestone and an antagonist, mifeprestone, were investigated in wildtype mice. The effect of progesterone was also investigated in mice lacking the classical progesterone receptor. RESULTS: Progesterone, as well as the more specific PR agonist promegestone, significantly reduced the CBF at concentrations of 10-100 nanomolar within 10-30 minutes. In the absence of progesterone, the PR antagonist mifeprestone had no effect on the ciliary beat frequency at a concentration of 1 micromolar. When ciliated cells were pre-incubated with 1 micromolar mifeprestone, addition of progesterone did not reduce the ciliary beat frequency. Accordingly, in ciliated cells from mice not expressing the classical progesterone receptor, exposure to 100 nanomolar progesterone did not reduce the ciliary beat frequency. CONCLUSIONS: This is the first study to provide comprehensive evidence that the classical progesterone receptor mediates the rapid reduction of the tubal ciliary beat frequency by progesterone.


Assuntos
Cílios/efeitos dos fármacos , Tubas Uterinas/efeitos dos fármacos , Progesterona/farmacologia , Receptores de Progesterona/fisiologia , Animais , Núcleo Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Óperon Lac/genética , Camundongos , Camundongos Endogâmicos C57BL , Progestinas/farmacologia , Promegestona/farmacologia , Receptores de Progesterona/agonistas , Receptores de Progesterona/antagonistas & inibidores
11.
J Biol Chem ; 286(50): 43091-102, 2011 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-22020934

RESUMO

The roles of progesterone (P(4)) and of progesterone receptor (PR) in development and pathogenesis of breast cancer remain unclear. In this study, we observed that treatment of T47D breast cancer cells with progestin antagonized effects of fetal bovine serum (FBS) to stimulate cell proliferation, whereas siRNA-mediated knockdown of endogenous PR abrogated progestin-mediated anti-proliferative effects. To begin to define mechanisms for the anti-proliferative action of P(4)/PR, we considered the role of MAPK phosphatase 1 (MKP-1/DUSP1), which catalyzes dephosphorylation and inactivation of MAPKs. Progestin treatment of T47D cells rapidly induced MKP-1 expression in a PR-dependent manner. Importantly, P(4) induction of MKP-1 was associated with reduced levels of phosphorylated ERK1/2, whereas siRNA knockdown of MKP-1 blocked progestin-mediated ERK1/2 dephosphorylation and repression of FBS-induced cell proliferation. The importance of PR in MKP-1 expression was supported by findings that MKP-1 and PR mRNA levels were significantly correlated in 30 human breast cancer cell lines. By contrast, no correlation was observed with the glucocorticoid receptor, a known regulator of MKP-1 in other cell types. ChIP and luciferase reporter assay findings suggest that PR acts in a ligand-dependent manner through binding to two progesterone response elements downstream of the MKP-1 transcription start site to up-regulate MKP-1 promoter activity. PR also interacts with two Sp1 sites just downstream of the transcription start site to increase MKP-1 expression. Collectively, these findings suggest that MKP-1 is a critical mediator of anti-proliferative and anti-inflammatory actions of PR in the breast.


Assuntos
Neoplasias da Mama/metabolismo , Fosfatase 1 de Especificidade Dupla/metabolismo , Receptores de Progesterona/metabolismo , Neoplasias da Mama/genética , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , Imunoprecipitação da Cromatina , Fosfatase 1 de Especificidade Dupla/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Immunoblotting , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Progesterona/farmacologia , Promegestona/farmacologia , Interferência de RNA , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Progesterona/agonistas , Receptores de Progesterona/genética
12.
Horm Behav ; 61(4): 598-604, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22366506

RESUMO

The influence of progesterone in the brain and on the behavior of females is fairly well understood. However, less is known about the effect of progesterone in the male system. In male rats, receptors for progesterone are present in virtually all vasopressin (AVP) immunoreactive cells in the bed nucleus of the stria terminalis (BST) and the medial amygdala (MeA). This colocalization functions to regulate AVP expression, as progesterone and/or progestin receptors (PR)s suppress AVP expression in these same extrahypothalamic regions in the brain. These data suggest that progesterone may influence AVP-dependent behavior. While AVP is implicated in numerous behavioral and physiological functions in rodents, AVP appears essential for social recognition of conspecifics. Therefore, we examined the effects of progesterone on social recognition. We report that progesterone plays an important role in modulating social recognition in the male brain, as progesterone treatment leads to a significant impairment of social recognition in male rats. Moreover, progesterone appears to act on PRs to impair social recognition, as progesterone impairment of social recognition is blocked by a PR antagonist, RU-486. Social recognition is also impaired by a specific progestin agonist, R5020. Interestingly, we show that progesterone does not interfere with either general memory or olfactory processes, suggesting that progesterone seems critically important to social recognition memory. These data provide strong evidence that physiological levels of progesterone can have an important impact on social behavior in male rats.


Assuntos
Progesterona/toxicidade , Reconhecimento Psicológico/efeitos dos fármacos , Comportamento Social , Tonsila do Cerebelo/fisiologia , Animais , Discriminação Psicológica/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Antagonistas de Hormônios/farmacologia , Masculino , Mifepristona/farmacologia , Congêneres da Progesterona/farmacologia , Promegestona/farmacologia , Desempenho Psicomotor/fisiologia , Ratos , Ratos Sprague-Dawley , Núcleos Septais/fisiologia , Olfato/efeitos dos fármacos , Vasopressinas/fisiologia
13.
Am J Obstet Gynecol ; 202(5): 455.e1-9, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20452487

RESUMO

OBJECTIVE: The purpose of this study was to evaluate cervical changes and delivery at term during pregnancy in rats after various progestin treatments. STUDY DESIGN: Pregnant rats were treated by various routes and vehicles with progesterone, 17-alpha-hydroxyprogesterone caproate (17P), R5020, and RU-486. Delivery time was determined and cervical ripening was assessed in vivo by collagen light-induced fluorescence. RESULTS: The cervix is rigid in the progesterone injection, 17P, and vaginal R5020 groups vs controls. Vaginal progesterone had no effect. RU-486 treatment softened the cervix during preterm delivery. Only subcutaneous injected progesterone, R5020 (subcutaneous and vaginal), and topical progesterone in sesame and fish oil inhibits delivery. Delivery is not changed by subcutaneous injection of 17P, vaginal progesterone, oral progesterone, and topical progesterone in Replens (Crinone; Columbia Labs, Livingston, NJ). CONCLUSION: Inhibition of cervical ripening and delivery by progestins depends on many factors that include their properties, the route of administration, and the vehicle. This study suggests reasons that the present treatments for preterm labor are not efficacious.


Assuntos
Maturidade Cervical/efeitos dos fármacos , Progestinas/farmacologia , 17-alfa-Hidroxiprogesterona/farmacologia , Animais , Feminino , Óleos de Peixe , Lipídeos , Gravidez , Nascimento Prematuro/prevenção & controle , Progesterona/administração & dosagem , Progesterona/farmacologia , Promegestona/farmacologia , Ratos , Ratos Sprague-Dawley , Óleo de Gergelim , Cremes, Espumas e Géis Vaginais
14.
Mol Endocrinol ; 23(1): 61-73, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19036901

RESUMO

The progesterone receptor (PR) plays a pivotal role in proper development and function of the mammary gland and has also been implicated in mammary tumorigenesis. PR is a ligand-activated transcription factor; however, relatively, little is known about its mechanisms of action at endogenous target promoters. The aim of our study was to identify a natural PR-responsive gene and investigate its transcriptional regulation in the mammary microenvironment. Our experiments revealed FKBP5 as a direct target of the PR, because it exhibited a rapid activation by progestin that was cycloheximide independent and correlated with recruitment of RNA polymerase II to the promoter. Site-directed mutagenesis and chromatin immunoprecipitation assays showed that progestin responsiveness is mediated through a composite element in the first intron, to which the PR binds concomitantly with GATA-2. Mutational analysis of the element revealed that the GATA-2 site is essential for progestin activation. Direct binding of PR to DNA contributes to the efficiency of activation but is not sufficient, suggesting that the receptor makes important protein-protein interactions as part of its mechanism of action at the FKBP5 promoter. Using chromatin immunoprecipitation assays we also determined that the intronic region is in communication with the promoter, probably via DNA looping. Time course analysis revealed a cyclical pattern of PR recruitment to the FKBP5 gene but a persistent recruitment to the mouse mammary tumor virus promoter, indicating that receptor cycling is a gene-specific phenomenon rather than a characteristic of the receptor itself. Our study offers new insight in the nature of PR-regulated transcription in mammary cancer cells.


Assuntos
Fator de Transcrição GATA2/metabolismo , Íntrons , Receptores de Progesterona/metabolismo , Proteínas de Ligação a Tacrolimo/genética , Animais , Sequência de Bases , Sítios de Ligação/genética , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , DNA de Neoplasias/genética , DNA de Neoplasias/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Células HeLa , Humanos , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Vírus do Tumor Mamário do Camundongo/genética , Camundongos , Modelos Biológicos , Mutagênese Sítio-Dirigida , Promegestona/farmacologia , Proteínas Recombinantes/genética
15.
Mol Reprod Dev ; 76(4): 367-78, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18932199

RESUMO

Members of the tumor necrosis factor (TNF)-receptor (R) family may be involved in the tissue remodeling that occurs in the primate corpus luteum (CL) during development and regression. As a first step towards addressing this issue, studies assessed TNF ligand-R expression and regulation in CL collected from monkeys during the early (ECL, Days 3-5), mid (MCL, Days 7-8), mid-late (MLCL, Days 10-11), late (LCL, Days 14-16), and very late (VLCL, menses) luteal phase of the menstrual cycle. CL were also collected after gonadotropin and/or steroid ablation and replacement (with hLH and the progestin R5020) for 3 days at mid-late luteal phase. TNF-alpha, -beta, FAS ligand (FASL), and TNF-R1 mRNA levels were two- to sixfold greater (P < 0.05) at the MLCL or LCL phase as compared to earlier (ECL, MCL). In contrast, TNF-R2 and FAS mRNA levels did not change during the luteal phase. Immunohistochemical staining for TNF-beta, TNF-R1, TNF-R2, FAS, and FASL was observed in luteal cells, whereas only TNF-beta staining was observed in endothelial cells. Several TNF-R components were influenced by LH and/or steroid ablation; notably, steroid ablation reduced (P < 0.05) luteal TNF-alpha, but not TNF-beta, mRNA levels, which was prevented by progestin treatment. In contrast, steroid ablation increased (P < 0.05) luteal cell immunostaining for FAS and FASL, which was reduced by progestin treatment. Thus, several members of the TNF R-ligand family are expressed in the primate CL in an LH- and/or progestin-dependent manner. Peak expression in the late luteal phase may signify a role for the TNF-R system in death receptor-mediated apoptosis during luteolysis.


Assuntos
Corpo Lúteo/metabolismo , Ciclo Menstrual/fisiologia , Receptores do Fator de Necrose Tumoral/metabolismo , Fatores de Necrose Tumoral/metabolismo , Abortivos Esteroides/farmacologia , Animais , Corpo Lúteo/citologia , Corpo Lúteo/efeitos dos fármacos , Di-Hidrotestosterona/análogos & derivados , Di-Hidrotestosterona/farmacologia , Feminino , Humanos , Macaca , Gravidez , Promegestona/farmacologia , RNA Mensageiro/metabolismo , Receptores do Fator de Necrose Tumoral/genética , Fatores de Necrose Tumoral/genética
16.
J Cell Biol ; 102(4): 1191-9, 1986 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-3958043

RESUMO

The progesterone receptor has been localized in the rabbit uterus by immunocytochemistry at the electron microscopic level, using monoclonal antibodies and the protein A-gold technique. The progesterone receptor in uterine stromal cells was mainly localized in the nucleus; however, a small fraction of antigen was present in the cytoplasm, where it was associated with the rough endoplasmic reticulum and with free ribosomes. The plasma membrane was not labeled. In the nucleus, the receptor was always associated with condensed chromatin or areas surrounding condensed chromatin, whereas the nuceolus was not labeled. In the chromatin, receptor distribution varied according to the hormonal state: in the absence of progesterone, the receptor was randomly scattered over the clumps of condensed chromatin; after administration of the progestin R5020, it was mainly detected in the border regions between condensed chromatin and nucleoplasm and, to a lesser extent, over dispersed chromatin in the nucleoplasm. These areas have been shown to be the most active sites of gene transcription.


Assuntos
Núcleo Celular/metabolismo , Norpregnadienos/farmacologia , Promegestona/farmacologia , Receptores de Progesterona/metabolismo , Útero/metabolismo , Animais , Anticorpos Monoclonais , Complexo Antígeno-Anticorpo/análise , Núcleo Celular/ultraestrutura , Feminino , Ouro , Histocitoquímica , Microscopia Eletrônica , Coelhos , Receptores de Progesterona/efeitos dos fármacos , Proteína Estafilocócica A , Útero/ultraestrutura
17.
Science ; 270(5240): 1354-7, 1995 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-7481822

RESUMO

A yeast two-hybrid system was used to identify a protein that interacts with and enhances the human progesterone receptor (hPR) transcriptional activity without altering the basal activity of the promoter. Because the protein stimulated transactivation of all the steroid receptors tested, it has been termed steroid receptor coactivator-1 (SRC-1). Coexpression of SRC-1 reversed the ability of the estrogen receptor to squelch activation by hPR. Also, the amino terminal truncated form of SRC-1 acted as a dominant-negative repressor. Together, these results indicate that SRC-1 encodes a coactivator that is required for full transcriptional activity of the steroid receptor superfamily.


Assuntos
Receptores de Progesterona/metabolismo , Receptores de Esteroides/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Ativação Transcricional , Sequência de Aminoácidos , Sequência de Bases , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Expressão Gênica , Células HeLa , Histona Acetiltransferases , Antagonistas de Hormônios/metabolismo , Antagonistas de Hormônios/farmacologia , Humanos , Mifepristona/metabolismo , Mifepristona/farmacologia , Dados de Sequência Molecular , Coativador 1 de Receptor Nuclear , Promegestona/farmacologia , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Transcrição/genética , Transfecção
18.
Toxicol Lett ; 186(2): 123-9, 2009 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-19429233

RESUMO

Specific molecular events, characteristic of each cell-cycle phase may have direct effect to the functionality of nuclear receptors. Based on this understanding, the evaluation of lipophilic chemicals at the different cell-cycle phases is significant and should be considered. In order to achieve the aim of performing large-scale dose-response analysis on the effects of lipophilic chemicals at the different cell-cycle phases, a stable, sensitive and highly selective human progesterone receptor (hPR) expressing HeLa reporter cell line, hPRLuc-20, was established. Upon the establishment of the hPRLuc-20 cells, they were synchronized to the G(1), S and G(2) phases and treated with progesterone (PROG) and promegestone (R5020). The cells successfully showed that at the different cell-cycle phase, both agonists resulted in different cellular responses. The differences in response supports that hPR expressed within the hPRLuc-20 cells do respond in a cell-cycle dependent manner, thus showing the cells' compatibility in large-scale dose-response analyses of chemicals. It is hopeful that the advanced application of the hPRLuc-20 cells could contribute to provide fundamental hints to further understand the function of hPR, and provide key observations to elucidate the nature of these chemicals with hPR, its corresponding co-regulators and transcription factors.


Assuntos
Receptores de Progesterona/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Relação Dose-Resposta a Droga , Citometria de Fluxo , Genes Reporter/genética , Células HeLa , Humanos , Lipídeos/química , Luciferases/biossíntese , Progesterona/farmacologia , Progestinas/farmacologia , Promegestona/farmacologia , Solubilidade
19.
Mol Endocrinol ; 22(4): 823-37, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18202149

RESUMO

Human progesterone receptors (PR) rapidly activate cytosolic signaling pathways, in addition to their classical function as ligand-activated transcription factors. Using ER+/PR-B+ T47D breast cancer cells, we probed the role of progestin-stimulated rapid PR signaling in the transcriptional regulation of target genes involved in breast cancer cell proliferation. Epidermal growth factor receptor (EGFR) was rapidly activated after a 10-min treatment with R5020. Progestin induced EGFR-, c-Src-, and MAPK-dependent phosphorylation of PR-B on the MAPK consensus site, Ser345. Ser345-phosphorylated PR-B receptors strongly associated with specificity protein 1 (Sp1) transcription factors to regulate PR cell cycle (p21) and growth-promoting (EGFR) target genes whose promoters lack canonical progesterone response element sequences. Inhibitors of EGFR, c-Src, or MAPK activities blocked PR tethering to Sp1 and progestin-stimulated S-phase entry. Mutant PR-B receptors defective for c-Src binding (mPro) were not phosphorylated on Ser345 in response to progestin and failed to interact with Sp1. Hormone-induced complexes containing Sp1 and wild-type PR-B, but not S345A or mPro PR-B, were recruited to Sp1 sites within the endogenous p21 promoter. Progestin-induced S-phase entry was attenuated in T47D cells containing wild-type PR-B and treated with EGFR, c-Src, or MAPK kinase inhibitors or in T47D cells stably expressing mPro or mutant DNA-binding domain PR-B. In sum, rapid progestin-activated PR signaling leads to PR Ser345 phosphorylation and tethering to Sp1. These events are critical for progestin-stimulated regulation of Sp1 target genes and breast cancer cell proliferation. Our data demonstrate the therapeutic potential for PR-targeted breast cancer treatment by exploiting multiple nodes along the PR signaling pathway, including PR-B, EGFR, c-Src, MAPK, or Sp1.


Assuntos
Receptores de Progesterona/metabolismo , Serina/metabolismo , Transdução de Sinais/fisiologia , Fator de Transcrição Sp1/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Imunoprecipitação da Cromatina , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Citometria de Fluxo , Humanos , Immunoblotting , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Modelos Biológicos , Fosforilação/efeitos dos fármacos , Progestinas/farmacologia , Promegestona/farmacologia , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Receptores de Progesterona/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fator de Transcrição Sp1/genética , Transcrição Gênica
20.
Climacteric ; 12(1): 80-7, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18979300

RESUMO

OBJECTIVES: The effect of the new progestogen drospirenone on biochemical markers in terms of cardiovascular effects was investigated in the presence and absence of aldosterone and compared to progesterone and the progestogens medroxyprogesterone acetate (MPA) and promegestone (R5020), and the antimineralocorticoid spironolactone. METHODS: Human female aortic endothelial cells were used for the experiments. The progestogens were tested alone at 0.1 and 10 micromol/l and in combination with aldosterone at a concentration of 10 micromol/l. The adhesion molecule E-selectin, the chemokine monocyte attracting protein-1 (MCP-1) and plasminogen activator inhibitor-1 (PAI-1) were chosen as markers. RESULTS: In combination with aldosterone, spironolactone, drospirenone, progesterone and R5020 were able to inhibit the aldosterone-induced increase in MCP-1 concentration, the effect being greatest for spironolactone. In contrast, MPA did not show any significant effect. For E-selectin, similar results were found; however, R5020 and MPA were not able to act antagonistically. Spironolactone, drospirenone and progesterone were able to significantly reduce the aldosterone-induced stimulation of PAI-1. For MPA and R5020, no significant effect was found. CONCLUSIONS: The new progestogen drospirenone seems to have favorable effects on the cardiovascular system due to its antimineralocorticoid property. Clinical studies must prove the results of this in vitro experiment.


Assuntos
Androstenos/farmacologia , Biomarcadores/análise , Sistema Cardiovascular/efeitos dos fármacos , Células Endoteliais/química , Antagonistas de Receptores de Mineralocorticoides/farmacologia , Adolescente , Aldosterona/farmacologia , Aorta , Cardiotônicos , Células Cultivadas , Quimiocina CCL2/análise , Vasos Coronários , Selectina E/análise , Células Endoteliais/efeitos dos fármacos , Feminino , Humanos , Inibidor 1 de Ativador de Plasminogênio/análise , Progesterona/farmacologia , Congêneres da Progesterona/farmacologia , Promegestona/farmacologia , Espironolactona/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA