Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Gynecol Oncol ; 138(2): 238-45, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26013696

RESUMO

OBJECTIVES: The study objectives were to determine baseline endometrial histology in morbidly obese women undergoing bariatric surgery and to assess the surgical intervention's impact on serum metabolic parameters, quality of life (QOL), and weight. METHODS: Women undergoing bariatric surgery were enrolled. Demographic and clinicopathologic data, serum, and endometrium (if no prior hysterectomy) were collected preoperatively and serum collected postoperatively. Serum global biochemical data were assessed pre/postoperatively. Welch's two sample t-tests and paired t-tests were used to identify significant differences. RESULTS: Mean age of the 71 women enrolled was 44.2 years, mean body mass index (BMI) was 50.9 kg/m(2), and mean weight loss was 45.7 kg. Endometrial biopsy results: proliferative (13/30; 43%), insufficient (8/30; 27%), secretory (6/30; 20%) and hyperplasia (3/30; 10%-1 complex atypical, 2 simple). QOL data showed significant improvement in physical component scores (PCS means 33.9 vs. 47.2 before/after surgery; p<0.001). Twenty women underwent metabolic analysis which demonstrated significantly improved glucose homeostasis, improved insulin responsiveness, and free fatty acid levels. Significant perturbations in tryptophan, phenylalanine and heme metabolism suggested decreased inflammation and alterations in the intestinal microbiome. Most steroid hormones were not significantly impacted with the exception of decreased DHEAS and 4-androsten metabolites. CONCLUSIONS: Bariatric surgery is accompanied by an improved physical quality of life as well as beneficial changes in glucose homeostasis, insulin responsiveness, and inflammation to a greater extent than the hormonal milieu. The potential cancer protective effects of bariatric surgery may be due to other mechanisms other than simply hormonal changes.


Assuntos
Cirurgia Bariátrica , Carcinogênese/patologia , Hiperplasia Endometrial/patologia , Endométrio/patologia , Obesidade/patologia , Obesidade/cirurgia , Adulto , Idoso , Peso Corporal , Carcinogênese/metabolismo , Neoplasias do Endométrio/metabolismo , Neoplasias do Endométrio/patologia , Neoplasias do Endométrio/prevenção & controle , Endométrio/metabolismo , Feminino , Glucose/metabolismo , Humanos , Metabolismo dos Lipídeos , Pessoa de Meia-Idade , Obesidade/metabolismo , Qualidade de Vida , Adulto Jovem
2.
Clin Immunol ; 151(1): 66-77, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24525049

RESUMO

Females of child-bearing age are more resistant to infectious disease and have an increased risk of systemic lupus erythematosus (SLE). We hypothesized that estrogen-induced gene expression could establish an immunoactivated state which would render enhanced defense against infection, but may be deleterious in autoimmune development. Using peripheral blood mononuclear cells (PBMCs), we demonstrate enhanced responses with immunogen stimulation in the presence of 17ß-estradiol (E2) and gene array analyses reveal toll-like receptor 8 (TLR8) as an E2-responsive candidate gene. TLR8 expression levels are up-regulated in SLE and PBMCs stimulated with TLR8 agonist display a female sex-biased, E2-sensitive response. Moreover, we identify a putative ERα-binding region near the TLR8 locus and blocking ERα expression significantly decreases E2-mediated TLR8 induction. Our findings characterize TLR8 as a novel estrogen target gene that can lower the inflammatory threshold and implicate an IFNα-independent inflammatory mechanism that could contribute to higher SLE incidence in women.


Assuntos
Endossomos/efeitos dos fármacos , Estradiol/farmacologia , Receptor alfa de Estrogênio/imunologia , Leucócitos Mononucleares/efeitos dos fármacos , Lúpus Eritematoso Sistêmico/imunologia , Receptor 8 Toll-Like/imunologia , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Células Cultivadas , Endossomos/imunologia , Endossomos/metabolismo , Receptor alfa de Estrogênio/genética , Feminino , Regulação da Expressão Gênica , Humanos , Imidazóis/farmacologia , Fatores Imunológicos/farmacologia , Interferon-alfa/imunologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Fatores Sexuais , Transdução de Sinais , Receptor 8 Toll-Like/agonistas , Receptor 8 Toll-Like/genética
3.
Int J Endocrinol ; 2022: 7690166, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35586275

RESUMO

Estradiol action is mediated by estrogen receptors (ERs), a and ß. Estradiol binding initiates ER-mediated transcription and ER degradation, the latter of which occurs via the ubiquitin-proteasome pathway. Inhibition of proteasome activity prevents estradiol-induced ERα degradation and transactivation. In ER-positive GH3 cells (a rat pituitary prolactinoma cell line), forskolin, acting via protein kinase A (PKA), stimulates ERα transcriptional activity without causing degradation, and proteasome inhibition does not block forskolin-stimulated transcription. Forskolin also protects liganded ERα from degradation. In the current study, we first examined ERα and ERß transcriptional activity in ER-negative HT22 cells and found that forskolin stimulated ERα-, but not ERß-dependent transcription, through the ligand-binding domain (LBD). We also identified four mutations (L396R, D431Y, Y542F, and K534E/M548V) on the ERα LBD that selectively obliterated the response to forskolin. In GH3 cells, transfected ERα mutants and ERß were protected from degradation by forskolin. Ubiquitination of ERα and ERß was increased by forskolin or estradiol. ERα ubiquitination was diminished by a mutated ubiquitin (K48R) that prevents elongation of polyubiquitin chains for targeting the proteasome. Increased ERα ubiquitination was not affected by the deletion of the A/B domain but significantly diminished in the F domain deletion mutant. Our results indicate distinct and novel mechanisms for forskolin stimulation of ERα transcriptional activity and protection from ligand-induced degradation. It also suggests a unique mechanism by which forskolin increases unliganded and liganded ERα and ERß ubiquitination but uncouples them from proteasome-mediated degradation regardless of their transcriptional responses to forskolin.

4.
J Neurosci ; 29(17): 5616-27, 2009 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-19403828

RESUMO

Gonadotropin-releasing hormone (GnRH) neurons are critical to controlling fertility. In vivo, estradiol can inhibit or stimulate GnRH release depending on concentration and physiological state. We examined rapid, nongenomic effects of estradiol. Whole-cell recordings were made of GnRH neurons in brain slices from ovariectomized mice with ionotropic GABA and glutamate receptors blocked. Estradiol was bath applied and measurements completed within 15 min. Estradiol from high physiological (preovulatory) concentrations (100 pm) to 100 nm enhanced action potential firing, reduced afterhyperpolarizing potential (AHP) and increased slow afterdepolarization amplitudes (ADP), and reduced I(AHP) and enhanced I(ADP). The reduction of I(AHP) was occluded by previous blockade of calcium-activated potassium channels. These effects were mimicked by an estrogen receptor (ER) beta-specific agonist and were blocked by the classical receptor antagonist ICI182780. ERalpha or GPR30 agonists had no effect. The acute stimulatory effect of high physiological estradiol on firing rate was dependent on signaling via protein kinase A. In contrast, low physiological levels of estradiol (10 pm) did not affect intrinsic properties. Without blockade of ionotropic GABA and glutamate receptors, however, 10 pm estradiol reduced firing of GnRH neurons; this was mimicked by an ERalpha agonist. ERalpha agonists reduced the frequency of GABA transmission to GnRH neurons; GABA can excite to these cells. In contrast, ERbeta agonists increased GABA transmission and postsynaptic response. These data suggest rapid intrinsic and network modulation of GnRH neurons by estradiol is dependent on both dose and receptor subtype. In cooperation with genomic actions, nongenomic effects may play a role in feedback regulation of GnRH secretion.


Assuntos
Estradiol/administração & dosagem , Receptor alfa de Estrogênio/fisiologia , Receptor beta de Estrogênio/fisiologia , Hormônio Liberador de Gonadotropina/fisiologia , Neurônios/fisiologia , Animais , Relação Dose-Resposta a Droga , Esquema de Medicação , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/classificação , Receptor beta de Estrogênio/agonistas , Receptor beta de Estrogênio/classificação , Feminino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Neurônios/efeitos dos fármacos , Subunidades Proteicas/agonistas , Subunidades Proteicas/fisiologia , Receptores de Estrogênio/agonistas , Receptores de Estrogênio/classificação , Receptores de Estrogênio/fisiologia , Fatores de Tempo
5.
Mol Endocrinol ; 23(2): 237-50, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19095772

RESUMO

GnRH is the main modulator of LH secretion and transcription of the LH subunit genes in pituitary gonadotropes. The LHbeta gene is preferentially transcribed during pulsatile GnRH stimuli of one pulse/30 min and is thus carefully controlled by specific signaling pathways and transcription factors. We now show that GnRH-stimulated LHbeta transcription is also influenced by the ubiquitin-proteasome system. GnRH-stimulated activity of an LHbeta reporter gene was prevented by proteasome inhibitors MG-132 and lactacystin. Inhibition was not rescued by overexpression of two key transcription factors for LHbeta, early growth response-1 (Egr-1) and steroidogenic factor-1 (SF-1). Increased endogenous LHbeta transcription after GnRH treatment was also prevented by MG-132, as measured by primary transcript assays. To investigate possible mechanisms of LHbeta transcriptional inhibition by proteasome blockade, we employed chromatin immunoprecipitation to measure LHbeta promoter occupancy by transcription factors. Without GnRH, binding was low and unorganized. With GnRH, Egr-1 and SF-1 associations were stimulated, cyclic, and coincidental, with a period of approximately 30 min. MG-132 disrupted GnRH-induced Egr-1 and SF-1 binding and prevented phosphorylated RNA polymerase II association with the LHbeta promoter. Egr-1, but not SF-1, protein was induced by GnRH and accumulated with MG-132. Egr-1 and SF-1 were ubiquitinated in gonadotropes and ubiquitinated forms of these factors associated with the LHbeta promoter, suggesting their degradation may be key for LHbeta proteasome-dependent transcription. Together, these results demonstrate that degradation via the proteasome is vital to GnRH-stimulated LHbeta expression, and this occurs in part by allowing proper transcription factor associations with the LHbeta promoter.


Assuntos
Regulação da Expressão Gênica , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Luteinizante Subunidade beta/metabolismo , Regiões Promotoras Genéticas , Complexo de Endopeptidases do Proteassoma/metabolismo , Subunidades Proteicas/metabolismo , Animais , Linhagem Celular , Inibidores de Cisteína Proteinase/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Genes Reporter , Leupeptinas/metabolismo , Hormônio Luteinizante Subunidade beta/genética , Camundongos , Inibidores de Proteassoma , Subunidades Proteicas/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Transdução de Sinais/fisiologia , Fator Esteroidogênico 1/genética , Fator Esteroidogênico 1/metabolismo , Ubiquitina/metabolismo
6.
Steroids ; 74(7): 622-7, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18996136

RESUMO

Both steroids and growth factors stimulate proliferation of steroid-dependent tumor cells, and interaction between these signaling pathways occurs at several levels. Steroid receptors are classified as ligand-activated transcription factors, and steps by which they activate target gene transcription are well understood. Several steroid responses have now been functionally linked to other intracellular signaling pathways, including c-Src or tyrosine kinase receptors. Steroids such as 17beta-estradiol (E2), via binding to cytoplasmic or membrane-associated receptors, were also shown to rapidly activate intracellular signaling cascades such as ERK, PI3K and STATs. These E2-stimulated phosphorylations can then contribute to altered tumor cell function. ER-positive breast cancer cells, in which proliferation is stimulated by E2 and suppressed by antiestrogens, have been of particular interest in dissecting nuclear and cytoplasmic roles of estrogen receptors (ER). In some cell contexts, ER interacts directly with the intracellular tyrosine kinase c-Src and other cytoplasmic signaling and adaptor molecules, such as Shc, PI3K, MNAR, and p130 Cas. Although the hierarchy among these associations is not known, it is clear that c-Src plays a fundamental role in both growth factor and E2-stimulated cell growth, and this may also require other growth factor receptors such as those for EGF or IGF-1. STAT transcription factors represent one pathway to integrate E2 cytoplasmic and nuclear signaling. STAT5 is phosphorylated in the cytoplasm at an activating tyrosine in response to E2 or EGF, and then is translocated to the nucleus to stimulate target gene transcription. E2 stimulates recruitment of STAT5 and ER to the promoter of several proliferative genes, and STAT5 knockdown prevents recruitment of either protein to these promoters. STAT5 activation by E2 in breast cancer cells requires c-Src and EGF receptor, and inhibition of c-Src or EGFR, or knockdown of STAT5, prevents E2 stimulation of several genes and breast cancer cell proliferation. Hyperactivation of the growth factor receptor-c-Src pathway can in some contexts decrease growth responses to E2, or render cells and tumors resistant to suppressive actions of endocrine therapies. Crosstalk between growth factors and steroids in both the cytoplasm and nucleus may thus have a profound impact on complex biological processes such as cell growth, and may play a significant role in the treatment of steroid-dependent breast cancers.


Assuntos
Núcleo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Citoplasma/efeitos dos fármacos , Estrogênios/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Humanos
7.
Mol Endocrinol ; 22(12): 2741-2, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18845669

RESUMO

This year, in response to member input and suggestions to highlight the vibrant basic science of endocrinology, The Endocrine Society Annual Meeting (ENDO 08) introduced a new feature, The Year in Basic Science series. Among the many interests and strengths of our members, three broad areas were chosen for initial consideration: nuclear receptors, kinase signaling, and hormones and cancer. Speakers were invited to present and discuss important publications during the past year between annual meetings (roughly June to June), and to put these findings into broad perspective for the endocrine community. Three distinguished researchers, Bert O'Malley, Tony Means, and Kate Horwitz, graciously agreed to participate in the inaugural venture, and this series of articles is based on their presentations at ENDO 08. Each individual approached this somewhat daunting task slightly differently. However, all three observed important and often common themes that ultimately link current basic molecular findings to broad translational and clinical problems, including metabolism and energy balance, neuronal migration and synapse formation, long-term memory formation, and endocrine pathology in cancer, reproduction and osteoporosis.


Assuntos
Pesquisa Biomédica/tendências , Endocrinologia/tendências , Animais , Congressos como Assunto , Endocrinologia/métodos , Hormônios/fisiologia , Humanos , Camundongos , Camundongos Knockout , Neoplasias/etiologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Transdução de Sinais/fisiologia
8.
Mol Endocrinol ; 22(8): 1781-96, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18550772

RESUMO

17beta-Estradiol (E2) acts through the estrogen receptor alpha (ERalpha) to stimulate breast cancer proliferation. Here, we investigated the functional relationship between ERalpha and signal transducer and activator of transcription (STAT)5b activity in ER+ MCF-7 and T47D human breast cancer cells after specific knockdown of STAT5b. STAT5b small interfering RNA (siRNA) inhibited E2-induced bromodeoxyuridine (BrdU) incorporation in both cell lines, as well as the E2-induced increase in MCF-7 cell number, cyclin D1 and c-myc mRNA, and cyclin D1 protein expression, indicating that STAT5b is required for E2-stimulated breast cancer proliferation. E2 treatment stimulated STAT5b tyrosine phosphorylation at the activating tyrosine Y699, resulting in increased STAT5-mediated transcriptional activity, which was inhibited by a Y669F STAT5b mutant. E2-induced STAT5-mediated transcriptional activity was inhibited by overexpressing a kinase-defective epidermal growth factor receptor (EGFR), or the EGFR tyrosine kinase inhibitor tyrphostin AG1478, indicating a requirement for EGFR kinase activity. Both E2-induced STAT5b tyrosine phosphorylation and STAT5-mediated transcription were also inhibited by the ER antagonist ICI 182,780 and the c-Src inhibitor PP2, indicating additional requirements for the ER and c-Src kinase activity. EGFR and c-Src kinase activities were also required for E2-induced cyclin D1 and c-myc mRNA. Together, these studies demonstrate positive cross talk between ER, c-Src, EGFR, and STAT5b in ER+ breast cancer cells. Increased EGFR and c-Src signaling is associated with tamoxifen resistance in ER+ breast cancer cells. Here we show that constitutively active STAT5b not only increased basal DNA synthesis, but also conferred tamoxifen resistance. Because STAT5b plays an integral role in E2-stimulated proliferation and tamoxifen resistance, it may be an effective therapeutic target in ER+ breast tumors.


Assuntos
Neoplasias da Mama/enzimologia , Receptores ErbB/metabolismo , Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais/efeitos dos fármacos , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Contagem de Células , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclina D1/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes Neoplásicos , Humanos , Fosforilação/efeitos dos fármacos , Fosfotirosina/metabolismo , Fator de Transcrição STAT5/genética , Tamoxifeno/farmacologia , Transcrição Gênica/efeitos dos fármacos
9.
Steroids ; 73(11): 1039-51, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18501937

RESUMO

The role of estrogen exposure in breast cancer risk is well-documented, and both estrogen synthesis and actions through the estrogen receptor (ER) have been targeted by therapies to control hormone-dependent breast cancer. The discovery of a second ER form and its therapeutic implications sparked great interest. Both the original ERalpha and the more recently identified ERbeta subtypes bind and respond similarly to many physiological and pharmacological ligands. However, differences in phytoestrogen binding have been noted, and subtype-specific ligands have been developed. Cell-based assays show that ERbeta and its variants are generally less active on gene transcription than ERalpha, and may influence ERalpha activity; however, both gene- and cell-specific responses occur, and nongenomic activities are less well explored. Specific ligands, and methods to disrupt or eliminate receptor subtype expression in animal and cell models, demonstrate that the ERs have both overlapping and distinct biological functions. Overall, in cell-based studies, ERalpha appears to play a predominant role in cell proliferation, and ERbeta is suggested to be antiproliferative. The potential for distinct populations of breast tumors to be identified based on ER subtype expression, and to exhibit distinct clinical behaviors, is of greatest interest. Several studies suggest that the majority of ER-positive tumors contain both subtypes, but that some tumors contain only ERbeta and may have distinct clinical behaviors and responses. Expression of ERbeta together with ERalpha favors positive responses to endocrine therapy in most studies, and additional studies to determine if the addition of ERbeta to ERalpha as a tumor marker is of clinical benefit are warranted. In contrast, the positive association between ERbeta and HER2 expression in high-grade ERalpha-negative breast cancer does not favor positive responses to endocrine therapy. Expression of ERbeta in specific clinical subpopulations, and the potential for therapies targeting ERbeta specifically, is discussed.


Assuntos
Neoplasias da Mama/metabolismo , Receptor beta de Estrogênio/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/genética , Feminino , Previsões , Humanos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Tamoxifeno/farmacologia , Tamoxifeno/uso terapêutico
10.
Mol Endocrinol ; 21(7): 1499-512, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17456797

RESUMO

The signaling pathways that are critical to the development and growth of breast cancer include those activated downstream of the estrogen receptor (ER) and the human epidermal growth factor receptor family. Many of these pathways, including the signal transducer and activator of transcription pathway, are common to both. The well-described genomic actions of ER involve its role as a transcription factor, either by binding directly to DNA through estrogen response elements, or by tethering to DNA through interaction with other proteins. Nongenomic signaling by the ER involves interaction with membrane-associated signaling proteins such as the c-Src tyrosine kinase and adapter proteins p130Cas and moderator of nongenomic activity of ER. Interactions with the signal transducer and activator of transcription pathway are important in both ER signaling pathways and are critical for estrogen-induced proliferation and tumorigenesis. These mechanisms of signaling cross talk and their role in resistance to antiestrogen therapies are discussed.


Assuntos
Neoplasias da Mama/metabolismo , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Resistencia a Medicamentos Antineoplásicos , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/metabolismo , Moduladores de Receptor Estrogênico/uso terapêutico , Feminino , Substâncias de Crescimento/genética , Substâncias de Crescimento/metabolismo , Humanos , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Modelos Biológicos , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Esteroides/metabolismo , Transcrição Gênica
11.
Cancer Res ; 66(14): 7007-15, 2006 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16849545

RESUMO

High expression of the adaptor molecule Cas has been linked to resistance to the antiestrogen tamoxifen, both in tissue culture and in human tumors. The aim of this study was to elucidate the mechanism(s) by which overexpression of Cas confers resistance to tamoxifen. Cas overexpression in MCF-7 breast cancer cells was shown to alleviate both tamoxifen-mediated growth inhibition and induction of apoptosis. This enhancement of cell proliferation/survival occurred in the absence of detectable effects on estrogen receptor (ER) transcriptional activity under conditions where tamoxifen was present, indicating that Cas-dependent tamoxifen resistance is not the result of a switch to an ER-negative phenotype or enhanced responses to the partial agonist activity of tamoxifen. Instead, we present evidence, suggesting that Cas promotes tamoxifen resistance by deregulation of alternative cell proliferation pathways, particularly those mediated through enhanced c-Src protein tyrosine kinase activity arising from Cas/c-Src interactions. Overexpression of Cas was found to drive endogenous c-Src into complex with Cas, a process that has been shown previously to cause up-regulation of c-Src tyrosine kinase activity. MCF-7 cells overexpressing Cas exhibited increased phosphorylation of two c-Src substrates, Tyr845 in the kinase domain of the epidermal growth factor receptor (EGFR) and signal transducer and activator of transcription (STAT) 5b. Importantly, Cas-dependent protection from the antiproliferative effects of tamoxifen was reversed by the expression of dominant inhibitory variants of these substrates (Y845F EGFR and COOH-terminally truncated STAT5b). Based on these findings, we suggest that the Cas/c-Src/EGFR/STAT5 signaling axis is a major regulator of tamoxifen-resistant breast cancer cell growth and survival.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Proteína Substrato Associada a Crk/metabolismo , Proteínas Tirosina Quinases/metabolismo , Fator de Transcrição STAT5/metabolismo , Tamoxifeno/farmacologia , Animais , Células COS , Proteína Tirosina Quinase CSK , Linhagem Celular Tumoral , Chlorocebus aethiops , Resistencia a Medicamentos Antineoplásicos , Humanos , Transdução de Sinais , Quinases da Família src
12.
Endocrinology ; 148(12): 6083-91, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17823254

RESUMO

Transcription of the LH subunit genes is stimulated by GnRH and may be modulated physiologically by steroids such as 17beta-estradiol (E). We found that E treatment amplified GnRH stimulation of the rat LHbeta and alpha-subunit promoters, and expression of the endogenous mRNA, in LbetaT2 gonadotrope cells 2- to 5-fold above GnRH alone. We examined gene expression in LbetaT2 cells after E and/or GnRH treatment, and found that E suppressed expression of transcription factor Zfhx1a, and enhanced GnRH stimulation of Egr-1 mRNA and protein. E effects were abolished in the presence of antiestrogen. Egr-1 is critical for LHbeta expression; however, the role of Zfhx1a, which binds to E-box sequences, was untested. We found E-box motifs in both the rat LHbeta (-381, -182, and -15 bp) and alpha-subunit (-292, -64, -58 bp) promoters. Zfhx1a overexpression suppressed basal and GnRH-stimulated activity of both promoters. Mutation of the alpha-subunit promoter E boxes at either -64 or -58 bp eliminated Zfhx1a suppression, whereas mutation of the -292 bp E box had no effect. Gel shift assays demonstrated that Zfhx1a bound to the -64 and -58, but not -292, bp E-box DNA. Similarly, mutation of LHbeta promoter E boxes at either -381 or -182, but not -15, bp reduced Zfhx1a suppression, correlating with binding of Zfhx1a. The -381 bp LHbeta E box overlaps with an Sp1 binding site in the distal GnRH-stimulatory region, and increased Sp1 expression overcame Zfhx1a suppression. Thus, one mechanism by which E may enhance GnRH-stimulated LH subunit promoter activity is through regulation of both activators and suppressors of transcription.


Assuntos
Estrogênios/farmacologia , Hormônio Liberador de Gonadotropina/farmacologia , Hormônio Luteinizante/genética , Regiões Promotoras Genéticas/genética , Fatores de Transcrição/genética , Transcrição Gênica/efeitos dos fármacos , Animais , Sítios de Ligação/genética , Linhagem Celular , Elementos E-Box/genética , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Immunoblotting , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo , Fatores de Transcrição/metabolismo
13.
Breast Cancer Res ; 9(3): 107, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17617929

RESUMO

In the previous issue of the journal, Lin and coworkers present data demonstrate that increased expression of estrogen receptor (ER)-beta in ER-alpha-positive breast cancer cells antagonizes a defined group of ER-alpha/estrogen stimulated genes that are involved in cell cycle regulation and DNA replication. Similar expression patterns for these genes were found human ER-alpha positive breast tumors expressing higher levels or ER-beta, and this correlated with better clinical outcome. The implications for these data, which suggest that ER-beta is a positive actor and diagnostic marker for therapeutic outcome, are discussed.


Assuntos
Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Receptor alfa de Estrogênio/fisiologia , Receptor beta de Estrogênio/fisiologia , Neoplasias da Mama/induzido quimicamente , Ciclo Celular , Replicação do DNA/genética , Receptor alfa de Estrogênio/antagonistas & inibidores , Estrogênios/efeitos adversos , Feminino , Humanos , Fatores de Risco , Transcrição Gênica
14.
Endocrinology ; 147(1): 543-51, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16210360

RESUMO

The estrogen receptor-alpha (ERalpha) pituitary-specific variant, TERP-1, is regulated dramatically by physiological status. We examined hormonal regulation of the TERP-1 promoter in transient transfection assays in GH3 somatolactotrope cells. We found that 17beta-estradiol (E2), genistein, androgen, pituitary adenylate cyclase-activating peptide, and forskolin (FSK) all stimulated TERP-1 promoter activity, whereas progesterone had no effect. ERalpha bound to a palindromic estrogen response element (ERE) and two half-site EREs; mutation of any of these sites decreased basal expression and completely obliterated E2 stimulation. In contrast, mutation of an activator protein-1 site decreased basal and FSK-stimulated promoter activity, but not E2 or androgen stimulation. The pure antiestrogen ICI 182,780 suppressed E2 and genistein, but not FSK or androgen, stimulation. Similarly, mutation of the ERE palindrome or half-site EREs suppressed promoter stimulation by E2 and genistein, but not by androgen or FSK. Because TERP-1 levels regulate ERalpha function on model promoters, we tested TERP-1 modulation of its own and other physiological promoters. TERP-1 suppressed basal and E2-stimulated expression of its own promoter. TERP-1 suppression required the ERE regions of the promoter, and the dimerization domain of TERP-1. TERP-1 overexpression also suppressed E2 stimulation of the progesterone receptor and prolactin promoters. Thus, estrogens, androgen, and FSK can stimulate TERP-1 promoter activity, and increased TERP-1 expression modulates E2 stimulation of physiological promoters. These data suggest that TERP-1 regulation may play a significant role in modifying pituitary ERalpha responses.


Assuntos
Androgênios/farmacologia , Colforsina/farmacologia , Receptor alfa de Estrogênio/genética , Estrogênios/farmacologia , Íntrons/genética , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Receptores de Estrogênio/genética , Animais , Sequência de Bases , Bovinos , Linhagem Celular , Primers do DNA , Sondas de DNA , Íntrons/efeitos dos fármacos , Cinética , Fígado/efeitos dos fármacos , Fígado/fisiologia , Ratos , Receptores de Estrogênio/efeitos dos fármacos , Transfecção
15.
Mol Endocrinol ; 19(11): 2660-70, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15976008

RESUMO

Breast cancer cell growth may be stimulated by 17beta-estradiol (E2) or growth factors like epidermal growth factor (EGF). However, tumors typically depend on only one of these pathways and may overexpress either estrogen receptor (ER) or EGF receptor (EGFR) and related family members. Tumors overexpressing EGFR are more aggressive than those expressing ER. Intracellular mediators of these growth-stimulatory pathways are not completely defined, but one potential common mediator of EGF and E2 signaling is the transcription factor signal transducer and activator of transcription 5 (STAT5). To investigate the role of STAT5 in potential crosstalk between E2 and EGF, MDA-MB231 and SKBr3 breast cancer cells, which are ER-negative and overexpress human EGF family receptors, were used. Introduction of ERalpha and treatment with E2 decreased EGF-induced tyrosine phosphorylation of STAT5b, basal and EGF-induced STAT5-mediated transcription, and EGF-stimulated DNA synthesis in these cells. Suppressive effects of E2-EpsilonRalpha were specific for STAT5, as EGF stimulation of MAPK was unaffected. Deletion/mutation analysis of ERalpha demonstrated that the DNA-binding domain was insufficient, and that the ligand-binding domain was required for these responses. ERalpha transcriptional activity was not necessary for suppression of STAT5 activity. Overexpression of c-Src did not prevent suppression of STAT5 activity by E2 and ERalpha. However, ERalpha did prevent basal increases in STAT5 activity with overexpressed c-Src. In the context of human EGF receptor family overexpression, E2-ER opposes EGF signaling by regulating STAT5 activity. STAT5 may be a crucial point of signaling for both E2 and growth factors in breast cancer cells, allowing targeted therapy for many types of breast tumors.


Assuntos
Neoplasias da Mama/metabolismo , Fator de Crescimento Epidérmico/antagonistas & inibidores , Receptores ErbB/metabolismo , Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Fator de Transcrição STAT5/antagonistas & inibidores , Neoplasias da Mama/genética , Bromodesoxiuridina/metabolismo , Ativação Enzimática , Receptor alfa de Estrogênio/genética , Genes Reporter , Humanos , Luciferases/análise , Luciferases/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais , Transcrição Gênica , Transfecção , Células Tumorais Cultivadas , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
16.
Oncogene ; 23(48): 7979-89, 2004 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-15489915

RESUMO

Both steroids and growth factors stimulate proliferation of steroid-dependent tumor cells, and interaction between these signaling pathways may occur at several levels. Steroid receptors are typically classified as ligand-activated transcription factors, and steps by which they bind ligand, dimerize, recruit coregulatory molecules, and activate target gene transcription are well understood. Several steroid responses are functionally linked to c-Src or tyrosine kinase receptors, and the physiological impact and the precise molecular pathways involved in these responses are under intensive investigation. Ligand-independent stimulation of steroid receptor-mediated transcription by growth factors is now believed to occur through activated protein kinases that phosphorylate the receptors and receptor coregulators. Recently, steroid hormones themselves have been shown to rapidly activate intracellular signaling cascades, via binding to cognate cytoplasmic or membrane-associated receptors. In some contexts, steroid receptors interact directly with c-Src and other cytoplasmic signaling molecules, such as Shc, PI3K, and p130 Cas. Crosstalk between growth factors and steroids in both the cytoplasm and nucleus could have profound impact on complex biological processes such as cell growth, and play a significant role in the treatment of steroid-dependent cancers. The potential roles of progesterone and estrogen receptors in this crosstalk are discussed in this review.


Assuntos
Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Quinases da Família src/metabolismo , Animais , Divisão Celular/fisiologia , Receptores ErbB/metabolismo , Estrogênios/metabolismo , Humanos , Progesterona/metabolismo , Receptores de Somatomedina/metabolismo
17.
Endocrinology ; 146(2): 751-9, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15528307

RESUMO

Estrogen receptor (ER)alpha is a ligand-inducible transcription factor that mediates the physiological effects of 17beta-estradiol (E2). In the uterus, E2 is involved in tissue growth, maintenance, and differentiation. Delta5ERalpha (Delta5) is an ERalpha variant protein expressed in uterine tumors but not in normal tissue. We examined the transcriptional activity of Delta5 and its modulation of human ERalpha basal and E2-stimulated activity in Ishikawa cells, an endometrial cancer cell line. In transient transfection assays, Delta5 increased basal activity of an estrogen response element-containing promoter in the absence or presence of ERalpha but lessened stimulation by ERalpha and E2. Effects of Delta5 were not limited to model reporters, given that cyclin D1 and complement 3 promoters were similarly affected. Increases in basal transcription required dimerization and DNA binding of Delta5, whereas decreased E2 stimulation with ERalpha required only DNA binding. Decreased ligand stimulation was not unique to E2 but also applied to the selective ER modulators tamoxifen and genistein. However, promoter stimulation by epidermal growth factor is retained with Delta5. The ERalpha coactivator small nuclear ring finger protein is expressed in Ishikawa cells and uterine tumors, and it enhances effects of Delta5 alone and with ERalpha on basal activity of an estrogen response element reporter. Thus, in the presence of Delta5 plus ERalpha, there is a lower transcriptional response to E2 and SERMS, but stimulation by epidermal growth factor is retained. The expression of Delta5 in uterine carcinoma may provide a mechanism by which tumors could maintain expression of E2-responsive genes in the absence of E2.


Assuntos
Neoplasias do Endométrio , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Útero/fisiologia , Linhagem Celular Tumoral , Dimerização , Fator de Crescimento Epidérmico/metabolismo , Estradiol/metabolismo , Receptor alfa de Estrogênio/química , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Ligantes , Mutagênese Sítio-Dirigida , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas/fisiologia , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Serina/metabolismo , Útero/citologia
18.
Mol Endocrinol ; 18(5): 1263-76, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-14988433

RESUMO

GnRH controls expression of the LH subunit genes, alpha and LHbeta, with the LHbeta subunit regulated most dramatically. Two enhancer regions, distal and proximal, on the rat LHbeta gene promoter cooperate for full basal expression and GnRH stimulation. It has been hypothesized that the transcription factors binding to these regions, Sp1, Egr-1, and steroidogenic factor 1 (SF-1), may interact directly or indirectly via a coactivator. One such coactivator may be small nuclear RING finger protein (SNURF), which is expressed in pituitary tissue and the LbetaT2 gonadotrope cell line. In transfection experiments in LbetaT2 cells, SNURF stimulated basal expression of LHbeta and increased overall GnRH stimulation. SNURF specifically stimulated LHbeta, with no effect on the alpha-subunit promoter. SNURF interacts with Sp1 and SF-1, but not Egr-1, in pull-down experiments. Point mutations or deletions of SNURF functional domains demonstrated that Sp1 and SF-1 interactions with SNURF are required for SNURF stimulatory effects on the LHbeta promoter. Endogenous SNURF is associated with the LHbeta promoter on native chromatin, suggesting that it plays a physiological role in LHbeta gene expression. SNURF also binds the androgen receptor, and SNURF overexpression overcomes androgen suppression of GnRH-stimulated LHbeta but not alphasubunit promoter activity. SNURF mutations that disrupt Sp1 or SF-1 binding eliminate rescue by SNURF. We conclude that SNURF may mediate interactions between the distal and proximal GnRH response regions of the LHbeta promoter to stimulate transcription and can also protect the promoter from androgen suppression.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Luteinizante Subunidade beta/metabolismo , Proteínas Nucleares/metabolismo , Fator de Transcrição Sp1/metabolismo , Fatores de Transcrição/metabolismo , Animais , Células Cultivadas , Proteínas de Ligação a DNA/genética , Proteínas de Homeodomínio , Mutação/genética , Proteínas Nucleares/genética , Regiões Promotoras Genéticas/genética , Ratos , Receptores Androgênicos/metabolismo , Receptores Citoplasmáticos e Nucleares , Fator de Transcrição Sp1/genética , Fator Esteroidogênico 1 , Fatores de Transcrição/genética , Ubiquitina-Proteína Ligases
19.
PLoS One ; 10(1): e0116825, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25617744

RESUMO

Luteinizing hormone (LH) is synthesized and secreted throughout the reproductive cycle from gonadotrope cells in the anterior pituitary, and is required for steroidogenesis and ovulation. LH contains an α-subunit common with FSH, and a unique LHß subunit that defines biological activity. Basal LHß transcription is low and stimulated by hypothalamic GnRH, which induces synthesis of early growth response protein-1 (Egr1), and stimulates binding of transcription factors Egr1 and steroidogenic factor-1 (SF1) on the promoter. WT1 (Wilms tumor protein1) is a zinc finger transcription factor with an essential role in urogenital system development, and which regulates several reproductive genes via interactions with SF1 or binding to GC-rich elements such as Egr1 binding sites. We investigated a potential role for WT1 in LHß transcription in clonal mouse gonadotrope LßT2 cells. WT1 was present in LßT2 and mouse pituitary cells, and protein bound to the endogenous LHß promoter. Interestingly, mRNAs for WT1(+KTS), which contains a three amino-acid insertion between the 3rd and 4th zinc fingers, and the WT1 (-KTS) variant were both expressed at significant levels. WT1 mRNAs and protein were decreased approximately 50% by GnRH treatment, under conditions where Egr1 mRNA and protein, and LHß transcription, were stimulated. Decreasing expression of mRNA for WT1 (-KTS) decreased stimulation of LHß and Egr1 by GnRH, whereas decreasing both WT1 (-KTS) and (+KTS) increased endogenous LHß transcription, and prevented LHß but not Egr1 stimulation by GnRH, suggesting differing biological activities for the WT1 isoforms. Overexpression of WT1 showed that WT1(-KTS) enhanced LHß promoter GnRH stimulation 2-to-3-fold and required the 3'Egr1 site, but WT1(+KTS) repressed both basal and GnRH-stimulated LHß promoter activity by approximately 70%. Our data suggest that WT1 can modulate LHß transcription, with differential roles for the two WT1 variants; WT1 (-KTS) enhances and WT1 (+KTS) suppresses transcription.


Assuntos
Variação Genética , Hormônio Luteinizante Subunidade beta/genética , Transcrição Gênica , Proteínas WT1/genética , Proteínas WT1/metabolismo , Animais , Linhagem Celular , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/farmacologia , Masculino , Camundongos , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica/efeitos dos fármacos
20.
Endocrinology ; 144(6): 2409-16, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12746302

RESUMO

Calcium influx plays a critical role in GnRH regulation of rat LH subunit gene transcription, but the site(s) of action are undefined. We investigated the potential of GnRH acting through calcium to activate calcium/calmodulin-dependent protein kinase type II (Ca/CaMK II) in mouse gonadotrope-derived LbetaT2 cells. GnRH stimulated Ca/CaMK II beta subunit activity 3-fold 2 min after treatment and returned to control values by 45 min. The Ca/CaMK II response to GnRH was blocked by administration of the Ca/CaMK II-specific inhibitor, KN-93. The calcium channel activator Bay K 8644 stimulated a 3-fold increase in Ca/CaMK II activity, similar to GnRH. Blocking calcium influx with nimodipine or depleting intracellular calcium storage pools with thapsigargin each resulted in a partial suppression of GnRH-induced activation of Ca/CaMK II, and in combination, completely suppressed the Ca/CaMK II response to GnRH. KN-93 and nimodipine also suppressed alpha-subunit and LHbeta promoter responses to GnRH by 40-60%. LHbeta promoter constructs containing either proximal or proximal and distal GnRH-responsive regions were sensitive to inhibition. These data show for the first time that Ca/CaMK II activation plays an important role in the transmission of GnRH signals from the plasma membrane to the LH subunit genes.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Cálcio/metabolismo , Hormônio Liberador de Gonadotropina/farmacologia , Hormônio Luteinizante Subunidade beta/genética , Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/farmacologia , Animais , Benzilaminas/farmacologia , Agonistas dos Canais de Cálcio/farmacologia , Canais de Cálcio/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Subunidade beta do Hormônio Folículoestimulante/genética , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Camundongos , Fosforilação , Hipófise/citologia , Regiões Promotoras Genéticas/fisiologia , Sulfonamidas/farmacologia , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/fisiologia , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA