Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
2.
Cancer Res ; 74(15): 4024-9, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-25035389

RESUMO

Research advances build upon the validity and reproducibility of previously published data and findings. Yet irreproducibility in basic biologic and preclinical research is pervasive in both academic and commercial settings. Lack of reproducibility has led to invalidated research breakthroughs, retracted articles, and aborted clinical trials. Concerns and requirements for transparent, reproducible, and translatable research are accelerated by the rapid growth of "post-publication peer review," open access publishing, and data sharing that facilitate the identification of irreproducible data/studies; they are magnified by the explosion of high-throughput technologies, genomics, and other data-intensive disciplines. Collectively, these changes and challenges are decreasing the effectiveness of traditional research quality mechanisms and are contributing to unacceptable-and unsustainable-levels of irreproducibility. The global oncology and basic biologic research communities can no longer tolerate or afford widespread irreproducible research. This article discusses (i) how irreproducibility in preclinical research can ultimately be traced to an absence of a unifying life science standards framework, and (ii) makes an urgent case for the expanded development and use of consensus-based standards to both enhance reproducibility and drive innovations in cancer research.


Assuntos
Pesquisa Biomédica/normas , Projetos de Pesquisa/normas , Animais , Humanos , Reprodutibilidade dos Testes
3.
Mol Pharmacol ; 78(6): 1046-58, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20837678

RESUMO

The liver X receptors (LXRα and LXRß) are members of the nuclear receptor superfamily that function as key transcriptional regulators of a number of biological processes, including cholesterol homeostasis, lipid metabolism, and keratinocyte differentiation. Natural ligands that activate LXRs include oxysterol derivatives such as 25-hydroxycholesterol, 27-hydroxycholesterol, 22(R)-hydroxycholesterol, 20(S)-hydroxycholesterol, and 24(S),25-epoxycholesterol. Related oxysterols, such as 5α,6α-epoxycholesterol (5,6-EC) are present in a number of foods and have been shown to induce atherosclerosis in animal models. Intriguingly, these oxysterols have also been detected in atherosclerotic plaques. Using a variety of biochemical and cellular assays, we demonstrate that 5,6-EC is the first dietary modulator and an endogenous LXR ligand with cell and gene context-dependent antagonist, agonist, and inverse agonist activities. In a multiplexed LXR-cofactor peptide interaction assay, 5,6-EC induced the recruitment of a number of cofactor peptides onto both LXRα and LXRß and showed an EC(50) of approximately 2 µM in peptide recruitment. Furthermore, 5,6-EC bound to LXRα in a radiolabeled ligand displacement assay (EC(50) = 76 nM), thus demonstrating it to be one of the most potent natural LXRα ligands known to date. Analysis of endogenous gene expression in various cell-based systems indicated the potential of 5,6-EC to antagonize LXR-mediated gene expression. Furthermore, it also induced the expression of some LXR-responsive genes in keratinocytes. These results clearly demonstrate that 5,6-EC is an LXR modulator that may play a role in the development of lipid disorders, such as atherosclerosis, by antagonizing the agonistic action of endogenous LXR ligands.


Assuntos
Colesterol/análogos & derivados , Receptores Nucleares Órfãos/metabolismo , Animais , Linhagem Celular Tumoral , Células Cultivadas , Colesterol/química , Colesterol/metabolismo , Colesterol/fisiologia , Humanos , Receptores X do Fígado , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Nucleares Órfãos/agonistas , Receptores Nucleares Órfãos/deficiência , Ligação Proteica/fisiologia , Transporte Proteico/fisiologia
4.
J Biol Chem ; 285(22): 17054-64, 2010 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-20356837

RESUMO

Selective androgen receptor modulators (SARMs) are androgen receptor (AR) ligands that induce anabolism while having reduced effects in reproductive tissues. In various experimental contexts SARMs fully activate, partially activate, or even antagonize the AR, but how these complex activities translate into tissue selectivity is not known. Here, we probed receptor function using >1000 synthetic AR ligands. These compounds produced a spectrum of activities in each assay ranging from 0 to 100% of maximal response. By testing different classes of compounds in ovariectomized rats, we established that ligands that transactivated a model promoter 40-80% of an agonist, recruited the coactivator GRIP-1 <15%, and stabilized the N-/C-terminal interdomain interaction <7% induced bone formation with reduced effects in the uterus and in sebaceous glands. Using these criteria, multiple SARMs were synthesized including MK-0773, a 4-aza-steroid that exhibited tissue selectivity in humans. Thus, AR activated to moderate levels due to reduced cofactor recruitment, and N-/C-terminal interactions produce a fully anabolic response, whereas more complete receptor activation is required for reproductive effects. This bimodal activation provides a molecular basis for the development of SARMs.


Assuntos
Androgênios/metabolismo , Azasteroides/farmacologia , Antagonistas de Hormônios/farmacologia , Receptores Androgênicos/química , Transcrição Gênica , Animais , Azasteroides/química , Células COS , Linhagem Celular Tumoral , Química Farmacêutica/métodos , Chlorocebus aethiops , Desenho de Fármacos , Feminino , Humanos , Ligantes , Masculino , Modelos Biológicos , Estrutura Terciária de Proteína , Ratos , Receptores Citoplasmáticos e Nucleares/metabolismo , Esteroides/metabolismo , Ativação Transcricional
5.
Mol Pharmacol ; 77(5): 744-50, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20110405

RESUMO

One of the many harmful factors faced by the skin is solar UV radiation, which damages skin by inducing chronic low-grade inflammation through increased expression of proinflammatory cytokines, metalloproteinases (MMPs) and cyclooxygenase-2 (COX-2). Estrogen receptors (ERs) alpha and beta are ligand-dependent transcription factors that are expressed in skin, and an ERbeta agonist has previously shown efficacy in vivo in models of pain and inflammation. Because ERbeta does not carry the breast and uterine proliferation liabilities of ERalpha, we decided to explore the possibility of using ERbeta as a target for photoaging. We show that ERbeta-selective compounds suppressed the expression of cytokines and MMPs in activated keratinocytes and fibroblast-based in vitro models of photoaging. Furthermore, in activated dermal fibroblasts, ERbeta-selective compounds also inhibited COX-2. These activities of ERbeta ligands in skin cells correlated with the expression levels of ERbeta and showed reversal by treatment with a potent synthetic ER antagonist. Furthermore, the pharmacology of ERbeta-selective compound was observed in wild-type but not in skin cells obtained from ERbeta knockout mice. Finally, we demonstrate that a synthetic ERbeta agonist inhibited UV-induced photodamage and skin wrinkle formation in a murine model of photoaging. Therefore, the potential of an ERbeta ligand to regulate multiple pathways underlying the cause of photoaging suggests ERbeta to be a novel therapeutic target for the prevention and treatment of photoaging.


Assuntos
Receptor beta de Estrogênio/fisiologia , Envelhecimento/efeitos da radiação , Animais , Citocinas/genética , Receptor beta de Estrogênio/deficiência , Receptor beta de Estrogênio/efeitos dos fármacos , Receptor beta de Estrogênio/genética , Feminino , Fibroblastos/fisiologia , Humanos , Ligantes , Metaloproteinases da Matriz/genética , Camundongos , Camundongos Pelados , Camundongos Knockout , Envelhecimento da Pele/genética , Envelhecimento da Pele/fisiologia , Luz Solar/efeitos adversos , Raios Ultravioleta/efeitos adversos
6.
Mol Endocrinol ; 23(1): 74-85, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19036900

RESUMO

The use of estrogen-based therapies and the selective estrogen receptor (ER) modulator (SERM), raloxifene, which are approved for postmenopausal osteoporosis, is associated with side effects such as uterine/breast hyperproliferation, thromboembolism, and hot flashes. A combination of a new SERM, bazedoxifene (BZA), and Premarin (conjugated estrogens; CE) is under investigation to mitigate the estrogen/SERM side effects with promising results in Phase III clinical trials. To explore the mechanism of BZA/CE action, we investigated the recruitment of cofactor peptides to ERalpha by components of CE and a mixture containing the 10 major components of CE with or without three different SERMs. Here, we demonstrate differential recruitment of cofactor peptides to ERalpha by the individual CE components using a multiplex nuclear receptor-cofactor peptide interaction assay. We show that estrone and equilin are partial agonists in comparison with 17beta-estradiol in recruiting cofactor peptides to ERalpha. Further, CE was more potent than 17beta-estradiol in mediating ERalpha interaction with cofactor peptides. Interestingly, BZA was less potent than other SERMs in antagonizing the CE-mediated cofactor peptide recruitment to ERalpha. Finally, in accordance with these biochemical findings, 17beta-estradiol and CE, as well as SERM/CE combinations, showed differential gene regulation patterns in MCF-7 cells. In addition, BZA showed antagonism of a unique set of CE-regulated genes and did not down-regulate the expression of a number of CE-regulated genes, the expression of which was effectively antagonized by the other two SERMs. These results indicate that SERMs in combination with CE exhibit differential pharmacology, and therefore, combinations of other SERMs and estrogen preparations may not yield the same beneficial effects that are observed in clinic by pairing BZA with CE.


Assuntos
Estrogênios Conjugados (USP)/farmacologia , Indóis/farmacologia , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Sequência de Aminoácidos , Sítios de Ligação , Conservadores da Densidade Óssea/administração & dosagem , Conservadores da Densidade Óssea/efeitos adversos , Conservadores da Densidade Óssea/farmacologia , Linhagem Celular , Interações Medicamentosas , Quimioterapia Combinada , Estradiol/administração & dosagem , Estradiol/efeitos adversos , Estradiol/farmacologia , Receptor alfa de Estrogênio/química , Receptor alfa de Estrogênio/metabolismo , Estrogênios Conjugados (USP)/administração & dosagem , Estrogênios Conjugados (USP)/efeitos adversos , Feminino , Perfilação da Expressão Gênica , Humanos , Indóis/administração & dosagem , Indóis/efeitos adversos , Ligantes , Dados de Sequência Molecular , Peptídeos/genética , Peptídeos/metabolismo , Estrutura Terciária de Proteína , Moduladores Seletivos de Receptor Estrogênico/administração & dosagem , Moduladores Seletivos de Receptor Estrogênico/efeitos adversos , Ativação Transcricional/efeitos dos fármacos
7.
Steroids ; 73(9-10): 901-5, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18261753

RESUMO

Estrogens play a critical role in the regulation of cellular proliferation, differentiation, and apoptosis. Evidence indicates that this regulation is mediated by a complex interface of direct control of gene expression (so-called "genomic action") and by regulation of cell-signaling/phosphorylation cascades (referred to as the "non-genomic", or "extranuclear" action). However, the mechanisms of the non-genomic action of estrogens are not well defined. We have recently described the identification of a novel scaffold protein termed MNAR (modulator of non-genomic action of estrogen receptor), that couples conventional steroid receptors with extranuclear signal transduction pathways, thus potentially providing additional and tissue- or cell-specific level of steroid hormone regulation of cell functions. We have demonstrated that the MNAR is required for ER alpha (ERa) interaction with p60(src) (Src), which leads to activation of Src/MAPK pathway. Our new data also suggest that activation of cSrc in response to E2 leads to MNAR phosphorylation, interaction with p85, and activation of the PI3 and Akt kinases. These data therefore suggest that MNAR acts as an important scaffold that integrates ERa action in regulation of important signaling pathways. ERa non-genomic action has been suggested to play a key role in estrogen-induced cardio-, neuro-, and osteo-protection. Therefore, evaluation of the molecular crosstalk between MNAR and ERa may lead to development of functionally selective ER modulators that can separate between beneficial, prodifferentiative effects in bone, the cardiovascular system and the CNS and the "detrimental", proliferative effects in reproductive tissues and organs.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Estrogênios/metabolismo , Regulação Neoplásica da Expressão Gênica , Fosfatidilinositol 3-Quinases/metabolismo , Transativadores/fisiologia , Quinases da Família src/metabolismo , Domínio Catalítico , Linhagem Celular Tumoral , Proliferação de Células , Proteínas Correpressoras , Ativação Enzimática , Humanos , Fosforilação , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Transdução de Sinais , Relação Estrutura-Atividade , Transativadores/metabolismo , Fatores de Transcrição
8.
EMBO J ; 27(3): 535-45, 2008 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-18219273

RESUMO

Estrogen deficiency in menopause is a major cause of osteoporosis in women. Estrogen acts to maintain the appropriate ratio between bone-forming osteoblasts and bone-resorbing osteoclasts in part through the induction of osteoclast apoptosis. Recent studies have suggested a role for Fas ligand (FasL) in estrogen-induced osteoclast apoptosis by an autocrine mechanism involving osteoclasts alone. In contrast, we describe a paracrine mechanism in which estrogen affects osteoclast survival through the upregulation of FasL in osteoblasts (and not osteoclasts) leading to the apoptosis of pre-osteoclasts. We have characterized a cell-type-specific hormone-inducible enhancer located 86 kb downstream of the FasL gene as the target of estrogen receptor-alpha induction of FasL expression in osteoblasts. In addition, tamoxifen and raloxifene, two selective estrogen receptor modulators that have protective effects in bone, induce apoptosis in pre-osteoclasts by the same osteoblast-dependent mechanism. These results demonstrate that estrogen protects bone by inducing a paracrine signal originating in osteoblasts leading to the death of pre-osteoclasts and offer an important new target for the prevention and treatment of osteoporosis.


Assuntos
Osso e Ossos/metabolismo , Estrogênios/fisiologia , Proteína Ligante Fas/biossíntese , Proteína Ligante Fas/fisiologia , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Células 3T3 , Animais , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Técnicas de Cocultura , Proteína Ligante Fas/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoblastos/citologia , Osteoclastos/citologia , Receptor fas/fisiologia
9.
J Cell Physiol ; 213(3): 610-7, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17886255

RESUMO

By regulating activities and expression levels of key signaling molecules, estrogens control mechanisms that are responsible for crucial cellular functions. Ligand binding to estrogen receptor (ER) leads to conformational changes that regulate the receptor activity, its interaction with other proteins and DNA. In the cytoplasm, receptor interactions with kinases and scaffolding molecules regulate cell signaling cascades (extranuclear/nongenomic action). In the nucleus, estrogens control a repertoire of coregulators and other auxiliary proteins that are associated with ER, which in turn determines the nature of regulated genes and level of their expression (genomic action). The combination of genomic and nongenomic actions of estrogens ultimately confers the cell-type and tissue-type selectivity. Recent studies have revealed some important new insights into the molecular mechanisms underlying ER action, which may help to explain the functional basis of existing selective ER modulators (SERMs) and provide evidence into how ER might be selectively targeted to achieve specific therapeutic goals. In this review, we will summarize some new molecular details that relate to estrogen signaling. We will also discuss some new strategies that may potentially lead to the development of functionally selective ER modulators that can separate between the beneficial, prodifferentiative effects in bone, the cardiovascular system and the CNS as well as the "detrimental," proliferative effects in reproductive tissues and organs.


Assuntos
Estrogênios/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Antagonistas de Estrogênios/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Ligantes , Modelos Biológicos , Receptores de Estrogênio/metabolismo , Receptores de Estrogênio/fisiologia , Moduladores Seletivos de Receptor Estrogênico/farmacologia
10.
Mol Cell Biol ; 27(5): 1904-13, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17194752

RESUMO

Estrogen actions are mediated by a complex interface of direct control of gene expression (the so-called "genomic action") and by regulation of cell signaling/phosphorylation cascades, referred to as the "nongenomic," or extranuclear, action. We have previously described the identification of MNAR (modulator of nongenomic action of estrogen receptor) as a novel scaffold protein that regulates estrogen receptor alpha (ERalpha) activation of cSrc. In this study, we have investigated the role of MNAR in 17beta-estradiol (E2)-induced activation of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. Consistent with our previous results, a direct correlation was established between MNAR expression levels and E2-induced activation of PI3 and Akt kinases. Endogenous MNAR, ERalpha, cSrc, and p85, the regulatory subunit of PI3 kinase, interacted in MCF7 cells treated with E2. The interaction between p85 and MNAR required activation of cSrc and MNAR phosphorylation on Tyr 920. Consequently, the mutation of this tyrosine to alanine (Y920A) abrogated the interaction between MNAR and p85 and the E2-induced activation of the PI3K/Akt pathway, which was required for the E2-induced protection of MCF7 cells from apoptosis. Nonetheless, the Y920A mutant potentiated the E2-induced activation of the Src/MAPK pathway and MCF7 cell proliferation, as observed with the wild-type MNAR. These results provide new and important insights into the molecular mechanisms of E2-induced regulation of cell proliferation and apoptosis.


Assuntos
Estrogênios/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Receptores de Estrogênio/metabolismo , Alanina/metabolismo , Substituição de Aminoácidos , Animais , Apoptose/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Ativação Enzimática/efeitos dos fármacos , Estradiol/metabolismo , Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Feminino , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imunoprecipitação , Sistema de Sinalização das MAP Quinases , Modelos Biológicos , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/metabolismo , Transfecção , Tirosina/metabolismo , Quinases da Família src/metabolismo
11.
Mol Endocrinol ; 20(3): 516-33, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16223974

RESUMO

Estrogen receptor alpha (ERalpha) serine 118 (Ser118) phosphorylation modulates activation function-1 (AF1) function. Correct positioning of helix 12 promotes agonist-dependent recruitment of cyclin-dependent kinase-7 to catalyze this event. In this study we show robust cyclin-dependent kinase-7-independent, AF2 antagonist-induced Ser118 phosphorylation. Estradiol (E2) and ICI-182,780 (ICI-780) induce Ser118 phosphorylation of wild-type ERalpha and either of two helix 12 mutants, suggesting AF2-independent action, probably via shedding of 90-kDa heat shock protein. With E2 treatment, the predominantly nuclear, phosphorylated ERalpha in COS-1 cells is detergent soluble. Although levels of ICI-780-induced phosphorylation are profound, Ser118-phosphorylated ERalpha is aggregated over the nucleus or in the cytoplasm, fractionating with the cell debris and making detection in cleared lysates improbable. Selective ER modulators (SERMs) elicit a mixed response with phosphorylated ERalpha in both detergent-soluble and -insoluble compartments. Apparent ligand-induced loss of ERalpha protein from cleared lysates is thus due to ligand-induced redistribution into the pellet, not degradation. The COS-1 response to ICI-780 can be mimicked in MCF-7 cells treated with a proteasome inhibitor to block authentic ligand-induced degradation. With SERMs and antagonists, the magnitude of Ser118-phosphorylated receptor redistribution into the insoluble fraction of COS-1 cells correlates with the magnitude of authentic ERalpha degradation in MCF-7 cells. A strong inverse correlation with ligand-induced uterotropism in vivo (P < 0.0001) and direct correlation with AF2-independent transrepression of the matrix metalloprotease-1 promoter in endometrial cells in vitro are seen. These data suggest that ligand-induced Ser118 phosphorylation of ERalpha can be AF2 independent. Furthermore, they identify translocation of Ser118-phosphorylated ERalpha out of the nucleus, leading to cytoplasmic aggregation, as an antagonist pathway that may precede receptor degradation.


Assuntos
Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio/antagonistas & inibidores , Receptor alfa de Estrogênio/metabolismo , Animais , Benzoquinonas , Células COS , Chlorocebus aethiops , Quinases Ciclina-Dependentes/efeitos dos fármacos , Quinases Ciclina-Dependentes/metabolismo , Citoplasma/efeitos dos fármacos , Citoplasma/metabolismo , Endométrio/citologia , Endométrio/efeitos dos fármacos , Estradiol/análogos & derivados , Estradiol/farmacologia , Receptor alfa de Estrogênio/agonistas , Feminino , Fulvestranto , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Proteínas de Choque Térmico HSP90/efeitos dos fármacos , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Lactamas Macrocíclicas , Metaloproteinase 1 da Matriz/efeitos dos fármacos , Metaloproteinase 1 da Matriz/genética , Tamanho do Órgão/efeitos dos fármacos , Fosforilação , Regiões Promotoras Genéticas , Quinonas/farmacologia , Ratos , Ratos Sprague-Dawley , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Serina/metabolismo , Útero/efeitos dos fármacos , Quinase Ativadora de Quinase Dependente de Ciclina
12.
Endocrinology ; 146(11): 4568-76, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15994348

RESUMO

Dehydroepiandrosterone (DHEA) exhibits peak adrenal secretion in the fetus at term and around age 30 yr in the adult. Levels then progressively decline, which is associated with decreased levels of testosterone, dihydrotestosterone, and estrogen in peripheral tissues. DHEA supplementation in postmenopausal women increases bone formation and density, an effect mainly attributed to peripheral conversion to sex hormones. In this study, we tested DHEA for direct effects on the androgen (AR) and estrogen (ER) receptors. DHEA bound to AR with a Ki of 1 microM, which was associated with AR transcriptional antagonism on both the mouse mammary tumor virus and prostate-specific antigen promoters, much like the effects of bicalutamide. Unlike bicalutamide, DHEA stimulated, rather than inhibited, LNCaP cell growth, suggesting possible interaction with other hormone receptors. Indeed DHEA bound to ERalpha and ERbeta, with Ki values of 1.1 and 0.5 microM, respectively. Despite the similar binding affinities, DHEA showed preferential agonism of ERbeta with an EC50 of approximately 200 nm and maximal activation at 1 microM. With ERalpha we found 30-70% agonism at 5 microM, depending on the assay. Physiological levels of DHEA are approximately 30 nM and up to 90 nM in the prostate. DHEA at 30 nM is actually sufficient to activate ERbeta transcription to the same degree as estrogen at its circulating concentration, and additive effects are seen when the two were combined. Taken together, DHEA has the potential for physiologically relevant direct activation of ERbeta. With peak levels at term and age 30 yr, there is also a potential for antagonist effects on AR and partial agonism of ERalpha.


Assuntos
Antagonistas de Receptores de Andrógenos , Desidroepiandrosterona/farmacologia , Receptor alfa de Estrogênio/agonistas , Receptor beta de Estrogênio/agonistas , Animais , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Desidroepiandrosterona/metabolismo , Receptor beta de Estrogênio/genética , Humanos , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Transcrição Gênica/efeitos dos fármacos
13.
Bioorg Med Chem Lett ; 15(6): 1675-81, 2005 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15745820

RESUMO

The discovery, synthesis, and SAR of chromanes as ER alpha subtype selective ligands are described. X-ray studies revealed that the origin of the ER alpha-selectivity resulted from a C-4 trans methyl substitution to the cis-2,3-diphenyl-chromane platform. Selected compounds from this class demonstrated very potent in vivo antagonism of estradiol in an immature rat uterine weight assay, effectively inhibited ovariectomy-induced bone resorption in a 42 days treatment paradigm, and lowered serum cholesterol levels in ovx'd adult rat models. The best antagonists 8F and 12F also exhibited potent inhibition of MCF-7 cell growth and were shown to be estrogen receptor down-regulators (SERDs).


Assuntos
Cromanos/química , Cromanos/farmacologia , Receptor alfa de Estrogênio/metabolismo , Moduladores Seletivos de Receptor Estrogênico/química , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Animais , Sítios de Ligação , Linhagem Celular , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Ligantes , Modelos Químicos , Estrutura Molecular , Tamanho do Órgão , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade , Útero/efeitos dos fármacos
14.
Endocrinology ; 146(2): 564-78, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15498879

RESUMO

The androgen receptor (AR) is expressed in the uterus; however, the role of AR in female reproductive physiology is poorly understood. Here we examined the effects of androgens on uterine growth and gene expression in adult ovariectomized rats. Nonaromatizable AR-selective agonists potently stimulate hypertrophy and induce significant myometrial expansion distinct from that induced by 17beta-estradiol (E2). In the endometrium, androgens only modestly increase epithelial cell height and antagonize the trophic effects of E2. To identify underlying mechanisms, global changes in RNA levels 24 h after stimulation with E2 and 5alpha-dihydrotestosterone (DHT) were compared. A total of 491 genes were differentially expressed after E2 treatment, including key regulators of tissue remodeling, cell signaling, metabolism, and gene expression. Of the 164 transcripts regulated by DHT, 86% were also affected by E2, including trophic genes like IGF-I and epithelial secretory genes such as uterocalin. In estrogen receptor (ER)alpha knockout mice, DHT cannot induce uterine growth, suggesting a key role for ERalpha. However, DHT appears not to activate ERalpha directly because DHT induction of IGF-I is blocked by the AR antagonist bicalutamide, and multiple genes regulated directly by ERalpha were not induced by DHT. The similarity between estrogens and androgens instead could reflect general trophic signaling in reproductive tissues because 93 of the 503 genes regulated in the uterus are similarly affected during prostate growth. Thus androgens regulate the trophic environment and architecture of the rodent uterus via a gene expression program that is overlapping but distinct from the estrogen response.


Assuntos
Androgênios/metabolismo , Estradiol/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Útero/citologia , Útero/fisiologia , Androgênios/farmacologia , Animais , Diferenciação Celular , Divisão Celular , Di-Hidrotestosterona/metabolismo , Di-Hidrotestosterona/farmacologia , Estradiol/farmacologia , Feminino , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Genômica , Hipertrofia , Masculino , Camundongos , Tamanho do Órgão , Próstata/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores Androgênicos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Células Estromais/citologia , Células Estromais/fisiologia , Útero/crescimento & desenvolvimento
15.
Mol Endocrinol ; 18(8): 2011-23, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15131260

RESUMO

The androgen receptor (AR) is a member of the steroid receptor superfamily that plays critical roles in the development and maintenance of the male reproductive system and in prostate cancer. Actions of AR are controlled by interaction with several classes of coregulators. In this study, we have identified LATS2/KPM as a novel AR-interacting protein. Human LATS1 and LATS2 are tumor suppressors that are homologs of Drosophila warts/lats. The interaction surface of LATS2 is mapped to the central region of the protein, whereas the AR ligand binding domain is sufficient for this interaction. LATS2 functions as a modulator of AR by inhibiting androgen-regulated gene expression. The mechanism of LATS2-mediated repression of AR activity appears to involve the inhibition of AR NH2- and COOH-terminal interaction. Chromatin immunoprecipitation assays in human prostate carcinoma cells reveal that LATS2 and AR are present in the protein complex that binds at the promoter and enhancer regions of prostate-specific antigen, and overexpression of LATS2 results in a reduction in androgen-induced expression of endogenous prostate-specific antigen mRNA. Immunohistochemistry shows that LATS2 and AR are localized within the prostate epithelium and that LATS2 expression is lower in human prostate tumor samples than in normal prostate. The results suggest that LATS2 may play a role in AR-mediated transcription and contribute to the development of prostate cancer.


Assuntos
Antagonistas de Receptores de Andrógenos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Sítios de Ligação , Células COS , Chlorocebus aethiops , Elementos Facilitadores Genéticos/genética , Humanos , Ligantes , Masculino , Regiões Promotoras Genéticas/genética , Próstata/metabolismo , Próstata/patologia , Antígeno Prostático Específico/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Ligação Proteica , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/isolamento & purificação , Estrutura Terciária de Proteína , Receptores Androgênicos/química , Receptores Androgênicos/metabolismo , Ativação Transcricional , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/isolamento & purificação
16.
J Biol Chem ; 279(2): 1310-22, 2004 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-14576152

RESUMO

The androgen receptor (AR), when complexed with 5alpha-dihydrotestosterone (DHT), supports the survival and proliferation of prostate cells, a process critical for normal development, benign prostatic hypertrophy, and tumorigenesis. However, the androgen-responsive genetic pathways that control prostate cell division and differentiation are largely unknown. To identify such pathways, we examined gene expression in the ventral prostate 6 and 24 h after DHT administration to androgen-depleted rats. 234 transcripts were expressed significantly differently from controls (p < 0.05) at both time points and were subjected to extensive data mining. Functional clustering of the data reveals that the majority of these genes can be classified as participating in induction of secretory activity, metabolic activation, and intracellular signaling/signal transduction, indicating that AR rapidly modulates the expression of genes involved in proliferation and differentiation in the prostate. Notably AR represses the expression of several key cell cycle inhibitors, while modulating members of the wnt and notch signaling pathways, multiple growth factors, and peptide hormone signaling systems, and genes involved in MAP kinase and calcium signaling. Analysis of these data also suggested that p53 activity is negatively regulated by AR activation even though p53 RNA was unchanged. Experiments in LNCaP prostate cancer cells reveal that AR inhibits p53 protein accumulation in the nucleus, providing a post-transcriptional mechanism by which androgens control prostate cell growth and survival. In summary these data provide a comprehensive view of the earliest events in AR-mediated prostate cell proliferation in vivo, and suggest that nuclear exclusion of p53 is a critical step in prostate growth.


Assuntos
Di-Hidrotestosterona/farmacologia , Regulação da Expressão Gênica , Próstata/metabolismo , Receptores Androgênicos/metabolismo , Androgênios/farmacologia , Animais , Sítios de Ligação , Divisão Celular , Núcleo Celular/metabolismo , Sobrevivência Celular , Células Cultivadas , Biologia Computacional , DNA Complementar/metabolismo , Masculino , Microscopia de Fluorescência , Família Multigênica , Análise de Sequência com Séries de Oligonucleotídeos , Peptídeos/química , RNA Mensageiro/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fatores de Tempo , Proteína Supressora de Tumor p53/metabolismo
17.
Mol Endocrinol ; 17(11): 2329-39, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12893881

RESUMO

Cell programs such as proliferation and differentiation involve the sequential activation and repression of gene expression. Vitamin D, via its active metabolite 1,25-dihydroxyvitamin D [1,25-(OH)2D3)], controls the proliferation and differentiation of a number of cell types, including keratinocytes, by directly regulating transcription. Two classes of coactivators, the vitamin D receptor (VDR)-interacting proteins (DRIP/mediator) and the p160 steroid receptor coactivator family (SRC/p160), control the actions of nuclear hormone receptors, including the VDR. However, the relationship between these two classes of coactivators is not clear. Using glutathione-S-transferase-VDR affinity beads, we have identified the DRIP/mediator complex as the major VDR binding complex in proliferating keratinocytes. After the cells differentiated, members of the SRC/p160 family were identified in the complex but not major DRIP subunits. Both DRIP and SRC proteins were expressed in keratinocytes. DRIP205 expression decreased during differentiation, although SRC-3 levels increased. Both DRIP205 and SRC-3 potentiated vitamin D-induced transcription in proliferating cells, but during differentiation, DRIP205 was no longer effective. These results indicate that these two distinct coactivators are sequentially involved in vitamin D regulation of gene transcription during keratinocyte differentiation, suggesting that these coactivators are part of the means by which the temporal sequence of gene expression is regulated during the differentiation process.


Assuntos
Queratinócitos/citologia , Queratinócitos/metabolismo , Proteínas Nucleares/metabolismo , Receptores de Calcitriol/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional , Sequência de Aminoácidos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Células Cultivadas , Histona Acetiltransferases , Humanos , Recém-Nascido , Queratinócitos/efeitos dos fármacos , Masculino , Espectrometria de Massas , Complexo Mediador , Subunidade 1 do Complexo Mediador , Dados de Sequência Molecular , Peso Molecular , Coativador 1 de Receptor Nuclear , Ligação Proteica , Ativação Transcricional/efeitos dos fármacos , Vitamina D/farmacologia
18.
Cancer Res ; 63(2): 432-40, 2003 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-12543799

RESUMO

Loss of genetic material on chromosome 6 has been associated with progression of human melanomas. We showed previously that introducing chromosome 6 into metastatic human melanoma cell lines suppresses metastasis without affecting the ability of the hybrids to form progressively growing tumors. By subtractive hybridization comparing nonmetastatic chromosome 6-containing (neo6/C8161) versus parental (C8161) metastatic cells, the KISS1 metastasis suppressor gene was isolated. However, KISS1 mapped to chromosome 1q32. To identify upstream regulator(s) of (and downstream effectors of) KISS1, microarray hybridization comparing C8161 and neo6/C8161 variants was performed. TXNIP/VDUP1, a thioredoxin-binding protein, was expressed more highly in neo6/C8161 and in nonmetastatic melanomas. Increased TXNIP expression inhibited metastasis and up-regulated KISS1. Surprisingly, TXNIP also mapped to chromosome 1q. PCR karyotyping that refined the region on chromosome 6 identified CRSP3/DRIP130, a transcriptional coactivator, as a metastasis suppressor. CRSP3 transfectant cells had up-regulated KISS1 and TXNIP expression and were suppressed for metastasis. Quantitative real-time reverse-transcription PCR of clinical melanoma samples showed that loss of CRSP3 expression correlated with decreased KISS1 expression and increased metastasis. Thus, we implicated a specific gene on chromosome 6 in the etiology of melanoma metastasis and identified potential up-stream regulators of KISS1 and TXNIP.


Assuntos
Proteínas de Transporte/genética , Cromossomos Humanos Par 6/genética , Genes Supressores de Tumor , Melanoma/genética , Melanoma/secundário , Proteínas/genética , Tiorredoxinas/genética , Transativadores , Fatores de Transcrição/genética , Animais , Proteínas de Transporte/biossíntese , Mapeamento Cromossômico , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Kisspeptinas , Complexo Mediador , Melanoma/metabolismo , Camundongos , Camundongos Nus , Análise de Sequência com Séries de Oligonucleotídeos , Biossíntese de Proteínas , Tiorredoxinas/biossíntese , Fatores de Transcrição/biossíntese , Transfecção , Transplante Heterólogo , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor
19.
J Biol Chem ; 277(17): 14359-62, 2002 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-11893728

RESUMO

Two functionally distinct classes of coactivators are recruited by liganded estrogen receptor, the DRIP/Mediator complex and p160 proteins, although the relative dynamics of recruitment is unclear. Previously, we have shown a direct, estradiol-dependent interaction between the DRIP205 subunit of the DRIP complex and the estrogen receptor (ER) AF2 domain. Here we demonstrate the in vivo recruitment of other endogenous DRIP subunits to ER in response to estradiol treatment in MCF-7 cells. To explore the relationship between DRIP and p160 coactivators, we examined the kinetics of coactivator recruitment to the ER target promoter, pS2, by chromatin immunoprecipitation. We observed a cyclic association and dissociation of coactivators with the promoter, with recruitment of p160s and DRIPs occurring in opposite phases, suggesting an exchange between these coactivator complexes at the target promoter.


Assuntos
Proteínas Nucleares/metabolismo , Receptores de Estrogênio/fisiologia , Transativadores/metabolismo , Humanos , Cinética , Complexo Mediador , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA