Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
Exp Physiol ; 95(7): 819-28, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20360425

RESUMO

The ciliary beat frequency (CBF) of guinea-pig fimbria during the ovarian cycle was measured by video microscopy using a high-speed camera (500 Hz). In the follicular phase, with increasing concentrations of beta-oestradiol ([betaE(2)]) and a low concentration of progesterone ([PRG]), CBF increased from 13.5 to 16 Hz. In the ovulatory phase, with further increase of [betaE(2)], CBF decreased gradually from 16 to 13.5 Hz. In the early luteal phase, with low [PRG] and [betaE(2)], CBF increased to 17 Hz; however, in the middle luteal phase, with increasing [PRG], CBF decreased (12 Hz), and in the late luteal phase, with decreasing [PRG], CBF increased to 15 Hz. Then, in the resting phase, with low [betaE(2)] and [PRG], CBF decreased immediately to 14 Hz. The CBF of the fimbria was measured in guinea-pigs treated with beta-oestradiol benzoate (betaE(2)B) or medroxyprogesterone (mPRG). A low dose of betaE(2)B increased CBF to 14.5 Hz, whereas a high dose decreased it to 11 Hz. A betaE(2) receptor blocker, ICI-182,780, abolished the betaE(2)B-induced CBF changes and maintained CBF at 12.0 Hz. Medroxyprogesterone decreased CBF to 12.5 Hz, and mifepristone (a PRG receptor blocker) abolished the mPRG-induced CBF decrease and maintained CBF at 15 Hz. The addition of both blockers increased CBF to 18 Hz, suggesting that activation of betaE(2) or PRG receptors decreases the CBF of the fimbria. In conclusion, a moderate [betaE(2)] increase maintains a high CBF (15.5 Hz) in the follicular phase, and then further [betaE(2)] increase decreases CBF to 13.5 Hz in the ovulatory phase. In the early and late luteal phase, low [betaE(2)] and [PRG] increase CBF to 17 and 15 Hz, respectively, and in the middle luteal phase a high [PRG] decreases CBF (to 12 Hz). Thus, the CBF of the fimbria was controlled by signals via betaE(2) and PRG receptors in guinea-pigs.


Assuntos
Cílios/efeitos dos fármacos , Cílios/fisiologia , Estradiol/farmacologia , Ciclo Estral/fisiologia , Progesterona/farmacologia , Animais , Estradiol/análogos & derivados , Ciclo Estral/efeitos dos fármacos , Tubas Uterinas/fisiologia , Feminino , Fulvestranto , Cobaias , Medroxiprogesterona/farmacologia , Microscopia de Vídeo , Mifepristona/farmacologia , Ovulação/fisiologia , Receptores de Estradiol/fisiologia , Receptores de Progesterona/fisiologia
3.
Gen Comp Endocrinol ; 161(1): 58-61, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18952087

RESUMO

The orphan G protein coupled receptor, GPR30, has the characteristics of a high affinity, specific estrogen membrane receptor on Atlantic croaker oocytes and mediates estrogen inhibition of oocyte maturation in this perciform fish. In order to determine the broad applicability of these findings to other teleosts, similar experiments were conducted in a cyprinid fish, zebrafish, in the present study. GPR30 mRNA expression was detected in zebrafish oocytes but not in the ovarian follicular cells. Both spontaneous and 17, 20beta-dihyroxy-4-pregnen-3-one (DHP)-induced maturation of follicle-enclosed zebrafish oocytes was significantly decreased when they were incubated with either estradiol-17beta, or the GPR30 agonists, ICI 182 780 and tamoxifen, or with the GPR30 specific agonist G-1. On the other hand spontaneous oocyte maturation increased two-fold when zebrafish ovarian follicles were incubated with an aromatase inhibitor, ATD. Moreover, the stimulatory effects of ATD on germinal vesicle breakdown (GVBD) were partially reversed by co-treatment with 100 nM of E2 or G-1. These results suggest that endogenous estrogens acting through GPR30 are involved in maintaining meiotic arrest of zebrafish oocytes.


Assuntos
Estradiol/metabolismo , Oócitos/fisiologia , Receptores de Estradiol/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Androstatrienos/farmacologia , Animais , Inibidores da Aromatase/farmacologia , Ciclopentanos/farmacologia , Estradiol/análogos & derivados , Estradiol/farmacologia , Feminino , Fulvestranto , Hidroxiprogesteronas/farmacologia , Quinolinas/farmacologia , Receptores de Estradiol/agonistas , Receptores Acoplados a Proteínas G/agonistas , Tamoxifeno/farmacologia , Peixe-Zebra
4.
Physiol Rev ; 88(1): 91-124, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18195084

RESUMO

Estradiol is the most potent and ubiquitous member of a class of steroid hormones called estrogens. Fetuses and newborns are exposed to estradiol derived from their mother, their own gonads, and synthesized locally in their brains. Receptors for estradiol are nuclear transcription factors that regulate gene expression but also have actions at the membrane, including activation of signal transduction pathways. The developing brain expresses high levels of receptors for estradiol. The actions of estradiol on developing brain are generally permanent and range from establishment of sex differences to pervasive trophic and neuroprotective effects. Cellular end points mediated by estradiol include the following: 1) apoptosis, with estradiol preventing it in some regions but promoting it in others; 2) synaptogenesis, again estradiol promotes in some regions and inhibits in others; and 3) morphometry of neurons and astrocytes. Estradiol also impacts cellular physiology by modulating calcium handling, immediate-early-gene expression, and kinase activity. The specific mechanisms of estradiol action permanently impacting the brain are regionally specific and often involve neuronal/glial cross-talk. The introduction of endocrine disrupting compounds into the environment that mimic or alter the actions of estradiol has generated considerable concern, and the developing brain is a particularly sensitive target. Prostaglandins, glutamate, GABA, granulin, and focal adhesion kinase are among the signaling molecules co-opted by estradiol to differentiate male from female brains, but much remains to be learned. Only by understanding completely the mechanisms and impact of estradiol action on the developing brain can we also understand when these processes go awry.


Assuntos
Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Estradiol/fisiologia , Animais , Animais Recém-Nascidos , Encéfalo/metabolismo , Feminino , Masculino , Sistema Nervoso/embriologia , Sistema Nervoso/crescimento & desenvolvimento , Receptores de Estradiol/fisiologia , Caracteres Sexuais , Transdução de Sinais/fisiologia
5.
Endocrinology ; 147(9): 4056-66, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16809439

RESUMO

There is an urgent need to identify and develop a new generation of therapeutic agents and systemic therapies targeting the estradiol (E2)/estrogen receptor (ER) signaling in breast cancer. In this regard, new information on the mechanisms of E2/ER function and/or cross talk with other prosurvival cascades should provide the basis for the development of other ideal anti-E2 therapies with the intent to enhance clinical efficacy, reduce side effects or both. Our very recent assessment of the mechanisms by which cancer-associated increased lipogenesis and its inhibition alters the E2/ER signaling discovered that fatty acid synthase (FASN), the enzyme catalyzing the terminal steps in the de novo biosynthesis of long-chain fatty acids, differentially modulates the state of sensitivity of breast and endometrial cancer cells to E2-stimulated ER transcriptional activation and E2-dependent cell growth and survival: 1) pharmacological inhibition of FASN activity induced a dramatic augmentation of E2-stimulated ER-driven gene transcription, whereas interference (RNAi)-mediated silencing of FAS gene expression drastically lowered E2 requirements for optimal activation of ER transcriptional activation in breast cancer cells; conversely, pharmacological and RNAi-induced inhibition of FASN worked as an antagonist of E2- and tamoxifen-dependent ER transcriptional activity in endometrial adenocarcinoma cells; 2) pharmacological and RNAi-induced inhibition of FASN synergistically enhanced E2-mediated down-regulation of ER protein and mRNA expression in breast cancer cells, whereas specific FASN blockade resulted in a marked down-regulation of E2-stimulated ER expression in endometrial cancer cells; and 3) FASN inhibition decreased cell proliferation and cell viability by promoting apoptosis in hormone-dependent breast and endometrial cancer cells. In this review we propose that, through a complex mechanism involving the regulation of MAPK/ER cross talk as well as critical E2-related proteins including the Her-2/neu (erbB-2) oncogene and the cyclin-dependent kinase inhibitors p21(WAF1/CIP1) and p27(Kip1), a previously unrevealed connection exists between FASN and the genomic and nongenomic ER activities in breast and endometrial cancer cells. From a clinical perspective, we suggest that if chemically stable FASN inhibitors or cell-selective systems able to deliver RNAi targeting FASN gene demonstrate systemic anticancer effects of FASN inhibition in vivo, additional preclinical studies to characterize their anti-breast cancer actions should be of great interest as the specific blockade of FASN activity may also provide a protective means against endometrial carcinoma associated with tamoxifen-based breast cancer therapy.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias do Endométrio/tratamento farmacológico , Inibidores Enzimáticos/uso terapêutico , Ácido Graxo Sintases/antagonistas & inibidores , Moduladores Seletivos de Receptor Estrogênico/uso terapêutico , Estradiol/fisiologia , Ácido Graxo Sintases/genética , Ácido Graxo Sintases/fisiologia , Ácidos Graxos/metabolismo , Feminino , Regulação Enzimológica da Expressão Gênica , Humanos , RNA Interferente Pequeno/farmacologia , Receptores de Estradiol/fisiologia , Transdução de Sinais
6.
Endokrynol Pol ; 56(3): 322-6, 2005.
Artigo em Polonês | MEDLINE | ID: mdl-16350726

RESUMO

Classical action of steroid hormones, called genomic, includes binding to their intracellular receptor, require hours or days to occur and require transcriptional effects with subsequent modulation of protein expression. Some of the biological effects induced by steroids, and mainly by sex steroids, take place within seconds or few minutes, time far too fast to be due to the genomic changes. The rapid, nongenomic action of estradiol are attributed to membrane action, probably through variety of proteins present in cell membrane. The rapid effects of steroid hormones are manifold, ranging from activation of protein and tyrosine kinases, G proteins, and modulation of ion channels. The nongenomic way of action includes also non-direct control of processes of transcription and gene expression. There are at least three different way to interact with cell membrane. Steroids may change membrane fluidity, without binding to any known protein or receptor. Another way is allosteric modulation of non-specific for steroid hormones receptors, or structural and enzymatic protein present in cell membrane. Evidence suggests that the classical steroid receptors can be localized at the plasma membrane, triggering signals typical for G-proteins coupled receptors. Physiological significance of nongenomic action of steroids needs to be elucidated.


Assuntos
Receptores de Superfície Celular/fisiologia , Receptores de Estradiol/fisiologia , Animais , Sítios de Ligação , Membrana Celular/metabolismo , Estradiol , Humanos , Receptores Acoplados a Proteínas G/fisiologia
8.
J Neurosci Res ; 78(5): 603-24, 2004 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-15515048

RESUMO

The extensive literature and the work from our laboratory illustrate the large number of complex processes affected by estrogen that might contribute to the striking ability of 17beta-estradiol (E2) and its derivatives to inhibit clinical and histological signs of experimental autoimmune encephalomyelitis (EAE) in mice. These effects require sustained exposure to relatively low doses of exogenous hormone and offer better protection when initiated prior to induction of EAE. However, oral ethinyl estradiol (EE) and fluasterone, which lacks estrogenic side effects, could partially reverse clinical EAE when given after the onset of disease. The three main areas discussed in this review include E2-mediated inhibition of encephalitogenic T cells, inhibition of cell migration into central nervous system tissue, and neuroprotective effects that promote axon and myelin survival. E2 effects on EAE were mediated through Esr1 (alpha receptor for E2) but not Esr2 (beta receptor for E2), as were its antiinflammatory and neuroprotective effects. A novel finding is that E2 up-regulated the expression of Foxp3 and CTLA-4 that contribute to the activity of CD4+CD25+ Treg cells. The protective effects of E2 in EAE suggest its use as therapy for MS, although the risk of cardiovascular disease may complicate treatment in postmenopausal women. This risk could be minimized by using subpregnancy levels of exogenous E2 that produced synergistic effects when used in combination another immunoregulatory therapy. Alternatively, one might envision using EE or fluasterone metabolites alone or in combination therapies in both male and female MS patients.


Assuntos
Encefalomielite Autoimune Experimental/terapia , Estradiol/uso terapêutico , Esclerose Múltipla/terapia , Fármacos Neuroprotetores/uso terapêutico , Animais , Antígenos CD , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Antígeno CTLA-4 , Movimento Celular/efeitos dos fármacos , Sistema Nervoso Central/citologia , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Estradiol/análogos & derivados , Estradiol/química , Feminino , Fatores de Transcrição Forkhead , Humanos , Imunoterapia/métodos , Masculino , Camundongos , Camundongos Transgênicos , Modelos Imunológicos , Fármacos Neuroprotetores/química , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Receptores de Estradiol/fisiologia , Caracteres Sexuais , Linfócitos T/efeitos dos fármacos , Linfócitos T/fisiologia
9.
Biol Reprod ; 65(5): 1311-23, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11673245

RESUMO

All mammalian uteri contain endometrial glands that synthesize or transport and secrete substances essential for survival and development of the conceptus (embryo/fetus and associated extraembryonic membranes). In rodents, uterine secretory products of the endometrial glands are unequivocally required for establishment of uterine receptivity and conceptus implantation. Analyses of the ovine uterine gland knockout model support a primary role for endometrial glands and, by default, their secretions in peri-implantation conceptus survival and development. Uterine adenogenesis is the process whereby endometrial glands develop. In humans, this process begins in the fetus, continues postnatally, and is completed during puberty. In contrast, endometrial adenogenesis is primarily a postnatal event in sheep, pigs, and rodents. Typically, endometrial adenogenesis involves differentiation and budding of glandular epithelium from luminal epithelium, followed by invagination and extensive tubular coiling and branching morphogenesis throughout the uterine stroma to the myometrium. This process requires site-specific alterations in cell proliferation and extracellular matrix (ECM) remodeling as well as paracrine cell-cell and cell-ECM interactions that support the actions of specific hormones and growth factors. Studies of uterine development in neonatal ungulates implicate prolactin, estradiol-17 beta, and their receptors in mechanisms regulating endometrial adenogenesis. These same hormones appear to regulate endometrial gland morphogenesis in menstruating primates and humans during reconstruction of the functionalis from the basalis endometrium after menses. In sheep and pigs, extensive endometrial gland hyperplasia and hypertrophy occur during gestation, presumably to provide increasing histotrophic support for conceptus growth and development. In the rabbit, sheep, and pig, a servomechanism is proposed to regulate endometrial gland development and differentiated function during pregnancy that involves sequential actions of ovarian steroid hormones, pregnancy recognition signals, and lactogenic hormones from the pituitary or placenta. That disruption of uterine development during critical organizational periods can alter the functional capacity and embryotrophic potential of the adult uterus reinforces the importance of understanding the developmental biology of uterine glands. Unexplained high rates of peri-implantation embryonic loss in humans and livestock may reflect defects in endometrial gland morphogenesis due to genetic errors, epigenetic influences of endocrine disruptors, and pathological lesions.


Assuntos
Útero/embriologia , Animais , Endométrio/embriologia , Endométrio/fisiologia , Estradiol/fisiologia , Feminino , Humanos , Morfogênese , Prolactina/fisiologia , Receptores de Estradiol/fisiologia , Receptores da Prolactina/fisiologia , Útero/fisiologia
10.
Endocrinology ; 142(4): 1669-77, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11250949

RESUMO

Estradiol (E(2))-signaling is widely considered to be exclusively mediated through the transcription-regulating intracellular estrogen receptor (ER) alpha and ERbeta. The aim of this study was to investigate transcription-independent E(2)-signaling in mouse IC-21 macrophages. E(2) and E(2)-BSA induce a rapid rise in the intracellular free Ca(2+) concentration ([Ca(2+)](i)) of Fura-2 loaded IC-21 cells as examined by spectrofluorometry. These changes in [Ca(2+)](i) can be inhibited by pertussis toxin, but not by the ER-blockers tamoxifen and raloxifene. The E(2)-signaling initiated at the plasma membrane is mediated through neither ERalpha nor ERbeta, but rather through a novel G protein-coupled membrane E(2)-receptor as revealed by RT-PCR, flow cytometry, and confocal laser scanning microscopy. A special feature of this E(2)-receptor is its sequestration upon agonist stimulation. Sequestration depends on energy and temperature, and it proceeds through a clathrin- and caveolin-independent pathway.


Assuntos
Estradiol/fisiologia , Receptores de Superfície Celular/fisiologia , Receptores de Estradiol/fisiologia , Transdução de Sinais/fisiologia , Animais , Cálcio/metabolismo , Células Cultivadas , Estradiol/metabolismo , Citometria de Fluxo , Fluoresceína-5-Isotiocianato , Macrófagos/metabolismo , Camundongos , Microscopia Confocal , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Soroalbumina Bovina/metabolismo
11.
J Cell Physiol ; 186(3): 414-24, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11169981

RESUMO

The mechanism by which estradiol (E2) acts on cell proliferation is still unclear. In this paper, we report the results of a series of experiments in an attempt to elucidate the effector pathway(s) involved in coupling the E2 receptors binding to cellular growth response in leiomyoma cells (LSMC). Under conditions of E2-dependent growth, E2 treatment of LSMC triggers rapid and transient activation of the MAP-kinase pathway. Interestingly, we demonstrate that the early downstream signal transduction events determined by E2-stimulation in quiescent LSMC, including the rapid protein tyrosine phosphorylation of a subset of intracellular proteins, such GAP, PI-3-K, and PLCgamma, and the concomitant activation of ancillary protein kinases, are related to E2-induced PDGF secretion. Moreover, we identify the PDGF, alone or in association with other growth factors, as the main growth factor involved in the proliferation response of LSMC to E2 stimulation. The addition of neutralizing antibodies anti-PDGF was able to inhibit the mitogenic activity present in LSMC conditioned media samples. On the other hand, E2 did not affect the constitutive expression as well as the ligand affinity of PDGF receptors on LSMC plasmamembrane. Cell treatment with the antiestrogen ICI 182780 correlate both with a perturbation of E2-induced transductional circuit and with the disappearance of the mitogenic factor, PDGF, in LSMC conditioned media; the latter therefore, represents the main autocrine mediator of cell growth modulation, upregulated by E2 and down-regulated by antiestrogenic compound. Our experiments suggest that growth factor secretion is an initial and integral part of the signaling events mediated by the estradiol receptors, not related, at least in part, to E2 transcriptional modulation.


Assuntos
Estradiol/análogos & derivados , Estradiol/farmacologia , Leiomioma/patologia , Sistema de Sinalização das MAP Quinases/fisiologia , Neoplasias Uterinas/patologia , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular , Meios de Cultivo Condicionados , Moduladores de Receptor Estrogênico/farmacologia , Feminino , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fulvestranto , Humanos , Cinética , Leiomioma/fisiopatologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfotirosina/análise , Fator de Crescimento Derivado de Plaquetas/farmacologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Proteínas Proto-Oncogênicas c-raf/metabolismo , Receptores de Estradiol/fisiologia , Receptores do Fator de Crescimento Derivado de Plaquetas/fisiologia , Células Tumorais Cultivadas , Neoplasias Uterinas/fisiopatologia
12.
Bone ; 27(1): 41-6, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10865207

RESUMO

Although estrogen is known to induce new bone formation in the long bones of female mice, this response is only thought to occur following administration of high doses, suggesting that it may not be mediated by a conventional estrogen receptor. To address this question further, we first examined the stereospecificity of this response by comparing the potency of 17beta-estradiol (E(2)) in stimulating cancellous bone formation at the proximal tibial metaphysis of intact female mice with that of the relatively inactive stereoisomer, 17alpha-estradiol (alphaE(2)). We found that E(2) was significantly more potent than alphaE(2), as assessed by histomorphometry. To provide further evidence for an estrogen-receptor-mediated process, we examined whether E(2)-induced osteogenesis in intact female mice could be inhibited by the estrogen receptor antagonist, ICI 182,780 (ICI). Although ICI itself had no effect on histomorphometric indices of the proximal tibial metaphysis when given alone, it significantly inhibited the osteogenic response to E(2). Finally, we examined the dose dependency of E(2)-induced osteogenesis at the proximal tibial metaphysis in intact mice. We found that E(2) stimulated cancellous bone formation in a dose-dependent manner over a wide dose range (i. e., 1-4000 microg/kg per day), with significant increases observed at doses of 4 microg/kg per day and beyond. Our results raise the possibility that estrogen-induced osteogenesis in the mouse represents an estrogen-receptor-mediated response that is not confined solely to supraphysiological estrogen levels.


Assuntos
Estradiol/farmacologia , Osteogênese/efeitos dos fármacos , Osteogênese/fisiologia , Receptores de Estradiol/fisiologia , Animais , Relação Dose-Resposta a Droga , Feminino , Camundongos , Camundongos Endogâmicos CBA
13.
J Immunol ; 163(7): 3758-63, 1999 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-10490972

RESUMO

We tested the hypothesis that estrogen acutely stimulates constitutive NO synthase (cNOS) activity in human peripheral monocytes by acting on an estrogen surface receptor. NO release was measured in real time with an amperometric probe. 17beta-estradiol exposure to monocytes stimulated NO release within seconds in a concentration-dependent manner, whereas 17alpha-estradiol had no effect. 17beta-estradiol conjugated to BSA (E2-BSA) also stimulated NO release, suggesting mediation by a membrane surface receptor. Tamoxifen, an estrogen receptor inhibitor, antagonized the action of both 17beta-estradiol and E2-BSA, whereas ICI 182,780, a selective inhibitor of the nuclear estrogen receptor, had no effect. We further showed, using a dual emission microfluorometry in a calcium-free medium, that the 17beta-estradiol-stimulated release of monocyte NO was dependent on the initial stimulation of intracellular calcium transients in a tamoxifen-sensitive process. Leeching out the intracellular calcium stores abolished the effect of 17beta-estradiol on NO release. RT-PCR analysis of RNA obtained from the cells revealed a strong estrogen receptor-alpha amplification signal and a weak beta signal. Taken together, a physiological dose of estrogen acutely stimulates NO release from human monocytes via the activation of an estrogen surface receptor that is coupled to increases in intracellular calcium.


Assuntos
Cálcio/sangue , Estradiol/sangue , Líquido Intracelular/metabolismo , Monócitos/metabolismo , Óxido Nítrico/sangue , Receptores de Estrogênio/sangue , Estradiol/fisiologia , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Expressão Gênica , Humanos , Receptores de Estradiol/fisiologia , Receptores de Estrogênio/antagonistas & inibidores , Receptores de Estrogênio/biossíntese , Receptores de Estrogênio/genética , Tamoxifeno/farmacologia
14.
Neoplasma ; 46(2): 112-6, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10466435

RESUMO

Estradiol receptors are regarded to predict a likely success of hormonal therapeutic efforts and the prognosis of breast cancer patients. But today its prognostic importance is controversial, discussed as either reflecting intrinsic property of the tumor tissue or better therapeutic accessibility of receptor positive tumors. Moreover, the most important clinical prognosticators--tumor size and axillary lymph node involvement do not seem to be related to the estradiol receptor status. In our investigation, the length of disease free interval is similar in estradiol receptor positive and negative patients and in all sites of distant metastases, but it is significantly reduced if more than 4 axillary lymph nodes are involved. Post recurrence survival is significantly longer in estradiol receptor positive than negative patients and also in patients treated by tamoxifen containing therapies. Its length is independent of the number of axillary lymph node metastases and the type of distant metastases, with a tendency to be longer in estradiol receptor positive than negative patients. In addition, the overall survival is longer for estradiol receptor positive than negative patients and becomes reduced with more than 4 axillary lymph node metastases. Frequency of deaths in estradiol receptor positive patients is half that of negative subjects. Furthermore, the length of overall survival is independent on the type of distant metastases, with tendency to be longer in estradiol receptor positive than negative patients. Longest overall survival could be observed for estradiol receptor positive patients who got therapy regimens containing tamoxifen. The weak prognostic advantages of estradiol receptor positive patients are interpreted by estradiol receptors as intrinsic parameters of breast cancer tissue characterizing more its biological behavior than therapeutic accessibility.


Assuntos
Neoplasias da Mama/metabolismo , Recidiva Local de Neoplasia/metabolismo , Receptores de Estradiol/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/cirurgia , Quimioterapia Adjuvante , Terapia Combinada , Ciclofosfamida/administração & dosagem , Intervalo Livre de Doença , Doxorrubicina/administração & dosagem , Feminino , Fluoruracila/administração & dosagem , Humanos , Metástase Linfática , Metotrexato/administração & dosagem , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/tratamento farmacológico , Prognóstico , Receptores de Estradiol/fisiologia , Tamoxifeno/administração & dosagem
15.
Pathologe ; 20(1): 50-5, 1999 Jan.
Artigo em Alemão | MEDLINE | ID: mdl-10091232

RESUMO

The ovarian steroid hormones estradiol and progesterone exert their effect by interacting with their intracellular receptors, which, after ligand binding translocate to the nucleus and bind to the promoter regions of target genes. The consequence is a change in the transcription rate of the target genes, followed by a change in production of the corresponding proteins. Target genes of the sexual steroid hormones include cytokines and growth factors, among them CSF-1, TGF-beta and LIF. The rhythm and activity of steroidogenesis, receptor modulation and transcription are reflected by cycle-specific proliferation and differentiation processes in the endometrium. Quantitative and/or qualitative molecular endocrinology is of increasing interest for better definition of morphological changes, although, as yet, the pathological laboratory test is of much less practical consequence than a suspicious vaginal sonography. In spite of the high standard of ultrasound techniques, however, most cases with slightly increased endometrial thickness show histologically benign changes of the endometrium rather than endometrial precancer or cancer. This is especially true for perimenopausal women with no other clinical findings. Yet, the cancer risk is increased in women under tamoxifen therapy. Hence, as a rule, these cases, when endometrial thickness exceeds 5 mm, need a diagnostic biopsy or abrasio.


Assuntos
Hiperplasia Endometrial/genética , Neoplasias do Endométrio/genética , Endométrio/patologia , Neoplasias Hormônio-Dependentes/genética , Receptores de Estradiol/fisiologia , Receptores de Progesterona/fisiologia , Biópsia , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Hiperplasia Endometrial/patologia , Neoplasias do Endométrio/patologia , Feminino , Humanos , Neoplasias Hormônio-Dependentes/patologia , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/patologia , Prognóstico , Sensibilidade e Especificidade
16.
Can J Physiol Pharmacol ; 76(4): 381-6, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9795746

RESUMO

In previous studies, we have shown that the baroreflex control of heart rate is significantly attenuated in females compared with age-matched males. This study investigated the role of estrogen in the modulation of baroreflex function in conscious unrestrained rats. Baroreflex-mediated decreases in heart rate in response to increments in blood pressure evoked by phenylephrine were evaluated in conscious freely moving male and female Sprague-Dawley rats as well as in ovariectomized rats. The effect of a 2-day 17 beta-estradiol (50 micrograms.kg-1.day-1, s.c.) or vehicle treatment on baroreflex sensitivity was investigated in ovariectomized rats. Intravenous bolus doses of phenylephrine (1-16 micrograms/kg) elicited dose-dependent pressor and bradycardic responses in all groups of rats. Regression analysis of the baroreflex curves relating increments in blood pressure to the associated heart rate responses revealed a significantly (p < 0.05) smaller baroreflex sensitivity in female compared with male rats (-1.22 +/- 0.07 and -1.85 +/- 0.15 beats.min-1.mmHg-1, respectively), suggesting an attenuated baroreflex function in females. In age-matched ovariectomized rats, baroreflex sensitivity showed further reduction (-0.93 +/- 0.02 beats.min-1.mmHg-1). Treatment of ovariectomized rats with 17 beta-estradiol significantly (p < 0.05) enhanced the baroreflex sensitivity (-1.41 +/- 0.16 beats.min-1.mmHg-1) to a level that was slightly higher than that of sham-operated female rats. Furthermore, baroreflex sensitivity of ovariectomized estradiol-treated rats was not significantly different from that of age-matched male rats. The vehicle, on the other hand, had no effect on baroreflex sensitivity of ovariectomized rats. These data support our earlier findings that sexual dimorphism exists in baroreflex control of heart rate. More importantly, the present study provides experimental evidence that suggests a facilitatory role for estrogen in the modulation of baroreflex function.


Assuntos
Barorreflexo/efeitos dos fármacos , Estradiol/farmacologia , Frequência Cardíaca/efeitos dos fármacos , Receptores de Estradiol/fisiologia , Animais , Barorreflexo/fisiologia , Feminino , Frequência Cardíaca/fisiologia , Masculino , Ovariectomia , Fenilefrina/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Estradiol/antagonistas & inibidores , Fatores Sexuais
17.
FASEB J ; 12(13): 1341-8, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9761777

RESUMO

Impaired insulin secretion is a hallmark in both type I and type II diabetic individuals. Whereas type I (insulin-dependent diabetes mellitus) implies ss-cell destruction, type II (non-insulin dependent diabetes mellitus), responsible for 75% of diabetic syndromes, involves diminished glucose-dependent secretion of insulin from pancreatic beta-cells. Although a clear demonstration of a direct effect of 17beta-estradiol on the pancreatic ss-cell is lacking, an in vivo insulinotropic effect has been suggested. In this report we describe the effects of 17beta-estradiol in mouse pancreatic ss-cells. 17beta-Estradiol, at physiological concentrations, closes K(ATP) channels, which are also targets for antidiabetic sulfonylureas, in a rapid and reversible manner. Furthermore, in synergy with glucose, 17beta-estradiol depolarizes the plasma membrane, eliciting electrical activity and intracellular calcium signals, which in turn enhance insulin secretion. These effects occur through a receptor located at the plasma membrane, distinct from the classic cytosolic estrogen receptor. Specific competitive binding and localization of 17beta-estradiol receptors at the plasma membrane was demonstrated using confocal reflective microscopy and immunocytochemistry. Gaining deeper knowledge of the effect induced by 17beta-estradiol may be important in order to better understand the hormonal regulation of insulin secretion and for the treatment of NIDDM. receptor.


Assuntos
Estradiol/farmacologia , Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Proteínas de Membrana/efeitos dos fármacos , Canais de Potássio/metabolismo , Potássio/metabolismo , Receptores de Estradiol/efeitos dos fármacos , Transportadores de Cassetes de Ligação de ATP , Trifosfato de Adenosina/metabolismo , Animais , Ligação Competitiva , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Permeabilidade da Membrana Celular , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Hipoglicemiantes/farmacologia , Técnicas Imunoenzimáticas , Secreção de Insulina , Transporte de Íons/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Canais KATP , Masculino , Potenciais da Membrana/efeitos dos fármacos , Proteínas de Membrana/fisiologia , Camundongos , Microscopia Confocal , Microscopia de Fluorescência , Técnicas de Patch-Clamp , Canais de Potássio Corretores do Fluxo de Internalização , Receptores de Estradiol/fisiologia , Transdução de Sinais , Compostos de Sulfonilureia/farmacologia
18.
Proc Natl Acad Sci U S A ; 94(11): 5872-6, 1997 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-9159167

RESUMO

Cyclic ADP ribose (cADPR) has been shown to trigger Ca2+ release from intracellular stores through ryanodine receptor/channel. In our previous study we observed that all-trans-retinoic acid stimulates cADPR synthesis by ADP ribose cyclase (ADPR cyclase) in cultured epithelial cells. We have now investigated whether cADPR may play a signaling role in action of beta-estradiol (E2), an archetypal steroid superfamily hormone, upon its major target organ, uterus, in vivo. Administration of E2 to gonadectomized rats (0.2 mg/kg per day for 7 days) resulted in an approximately Delta + 300% increase of ADPR cyclase activity in extracts from uterus, but in liver, brain, or skeletal muscle ADPR cyclase was unchanged. Most of the E2-stimulated uterine ADPR cyclase was associated with membranes. The higher ADPR cyclase activity in response to E2 was due to the increase of VMAX without change in Km. Simultaneous administration of estrogen antagonist tamoxifen (8 mg/kg per day) with E2 (0.2 mg/kg per day) prevented an increase in ADPR cyclase. In uterine extracts from E2-treated rats, the rate of cADPR inactivation by cADPR hydrolase and the activity of NADase was increased, but to a much lesser degree than activity of ADPR cyclase. Our results indicate that E2, via action to its nuclear receptors in vivo, increases ADPR cyclase activity in uterus. We propose that some of the estrogen effects, and by extension the effects of other steroid superfamily hormones, upon specialized cellular functions and upon hormone-induced gene expression in target cells, are mediated by cADPR-Ca2+ release pathway.


Assuntos
Adenosina Difosfato Ribose/análogos & derivados , Antígenos CD , Estradiol/farmacologia , Útero/enzimologia , ADP-Ribosil Ciclase , ADP-Ribosil Ciclase 1 , Adenosina Difosfato Ribose/isolamento & purificação , Adenosina Difosfato Ribose/metabolismo , Animais , Antígenos de Diferenciação/metabolismo , Encéfalo/enzimologia , Cálcio/metabolismo , Núcleo Celular/fisiologia , Cromatografia Líquida de Alta Pressão , ADP-Ribose Cíclica , Feminino , Rim/enzimologia , Cinética , Fígado/enzimologia , Glicoproteínas de Membrana , Modelos Biológicos , Músculo Liso/enzimologia , N-Glicosil Hidrolases/metabolismo , NAD/metabolismo , NAD+ Nucleosidase/metabolismo , Ovariectomia , Óvulo/fisiologia , Paratireoidectomia , Ratos , Ratos Sprague-Dawley , Receptores de Estradiol/fisiologia , Ouriços-do-Mar , Sistemas do Segundo Mensageiro , Tamoxifeno/farmacologia , Tireoidectomia
19.
J Mol Endocrinol ; 19(3): 249-57, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9460646

RESUMO

In the present study, the induction of the phosphoinositide signal transduction pathway by 17 beta-oestradiol has been demonstrated in rat vaginal epithelial cells (VEC). We have shown an increase in the metabolism of phosphoinositol lipids by 3H-myoinositol incorporation as well as production of inositol phosphate in VEC in vivo and in vitro following oestradiol administration. Concomitant changes in intracellular calcium levels have also been observed under the influence of oestradiol. To rule out the effects of cytokines, parallel studies were performed using primary cultures of VEC. Oestradiol-induced calcium uptake was seen even in the presence of actinomycin D and cycloheximide which inhibit transcription and translation respectively. Calcium uptake was blocked by neomycin, an inhibitor of phosphoinositol lipid metabolism, and by the oestrogen receptor antagonist tamoxifen. Results suggest that oestradiol induces second messenger pathways in the VEC through specific receptors. Implications of these observations in cellular differentiation processes are discussed.


Assuntos
Células Epiteliais/efeitos dos fármacos , Estradiol/farmacologia , Fosfatidilinositóis/farmacologia , Transdução de Sinais/efeitos dos fármacos , Vagina/citologia , Animais , Cálcio/metabolismo , Cálcio/farmacocinética , Cálcio/farmacologia , Células Cultivadas , Células Epiteliais/citologia , Feminino , Inositol/metabolismo , Fosfatidilinositóis/metabolismo , Fosfolipídeos/metabolismo , Ratos , Ratos Wistar , Receptores de Estradiol/fisiologia , Sistemas do Segundo Mensageiro/fisiologia , Transdução de Sinais/fisiologia , Esteroides/farmacologia , Trítio , Vagina/efeitos dos fármacos
20.
Mol Cell Endocrinol ; 124(1-2): 71-7, 1996 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-9027326

RESUMO

The mRNA levels of LIV-1 and pS2, two estrogen-responsive genes, are increased by the agents, cholera toxin (CT) plus 3-isobutyl-l-methylxanthine (IBMX), which cause an increase in cAMP in MCF-7 human breast cancer cells. The simultaneous addition of estradiol and CT/IBMX results in a synergistic induction of the two mRNAs. The changes in mRNA reflect changes in transcription of the two genes. Interestingly, the addition of CT/IBMX to estradiol not only causes a greater increase in transcription rate but the increase is longer-lasting that seen with the hormone alone. Stimulation of mRNA levels by CT/IBMX, but not by estradiol, was prevented by cycloheximide. Stimulation by both estradiol and by CT/IBMX was prevented by the antiestrogen, ICI 164387. Transcription of LIV-1 and pS2 genes is by both estradiol and cAMP, via separate mechanisms both requiring the estrogen receptor.


Assuntos
AMP Cíclico/fisiologia , Estradiol/farmacologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Proteínas de Neoplasias/genética , Proteínas , 1-Metil-3-Isobutilxantina/farmacologia , Neoplasias da Mama , Carcinoma , Toxina da Cólera/farmacologia , Cicloeximida/farmacologia , Estradiol/análogos & derivados , Antagonistas de Estrogênios/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Inibidores de Fosfodiesterase/farmacologia , Alcamidas Poli-Insaturadas , Inibidores da Síntese de Proteínas/farmacologia , RNA Mensageiro/biossíntese , RNA Neoplásico/biossíntese , Receptores de Estradiol/fisiologia , Fator Trefoil-1 , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...