Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Endocrinology ; 141(8): 2982-94, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10919287

RESUMO

Mammary glands from the estrogen receptor-a knockout (alphaERKO) mouse do not undergo ductal morphogenesis or alveolar development. Disrupted ERalpha signaling may result in reduced estrogen-responsive gene products in the mammary gland or reduced mammotropic hormones that contribute to the alphaERKO mammary phenotype. We report that circulating PRL is reduced in the female alphaERKO mouse. Implantation of an age-matched, heterozygous ERalpha pituitary isograft under the renal capsule of 25-day-old or 12-week-old alphaERKO mice increased circulating PRL and progesterone levels, and induced mammary gland development. Grafted alphaERKO mice also possessed hypertrophied corpora lutea demonstrating that PRL is luteotropic in the alphaERKO ovary. By contrast, ovariectomy at the time of pituitary grafting prevented mammary gland development in alphaERKO mice despite elevated PRL levels. Hormone replacement using pellet implants demonstrated that pharmacological doses of estradiol induced limited mammary ductal elongation, and estradiol in combination with progesterone stimulated lobuloalveolar development. PRL alone or in combination with progesterone or estradiol did not induce alphaERKO mammary growth. Estradiol and progesterone are required for the structural development of the alphaERKO mammary gland, and PRL contributes to this development by inducing ovarian progesterone levels. Therefore, the manifestation of the alphaERKO mammary phenotype appears due to the lack of direct estrogen action at the mammary gland and an indirect contributory role of estrogen signaling at the hypothalamic/pituitary axis.


Assuntos
Glândulas Mamárias Animais/crescimento & desenvolvimento , Receptores de Estrogênio/deficiência , Animais , Corpo Lúteo/patologia , Di-Hidrotestosterona/farmacologia , Estradiol/administração & dosagem , Estradiol/farmacologia , Receptor alfa de Estrogênio , Feminino , Hipertrofia , Camundongos , Camundongos Knockout , Ovariectomia , Hipófise/transplante , Progesterona/administração & dosagem , Progesterona/sangue , Progesterona/farmacologia , Prolactina/sangue , Receptores de Estrogênio/genética , Receptores de Estrogênio/fisiologia
2.
Endocrinology ; 140(12): 5855-65, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10579351

RESUMO

Ovarian-derived estradiol plays a critical endocrine role in the regulation of gonadotropin synthesis and secretion from the hypothalamic-pituitary axis. In turn, several para/autocrine effects of estrogen within the ovary are known, including increased ovarian weight, stimulation of granulosa cell growth, augmentation of FSH action, and attenuation of apoptosis. The estrogen receptor-alpha (ERalpha) is present in all three components of the hypothalamic-pituitary-ovarian axis of the mouse. In contrast, estrogen receptor-beta (ERbeta) is easily detectable in ovarian granulosa cells but is low to absent in the pituitary of the adult mouse. This distinct expression pattern for the two ERs suggests the presence of separate roles for each in the regulation of ovarian function. Herein, we definitively show that a lack of ERalpha in the hypothalamic-pituitary axis of the ERalpha-knockout (alphaERKO) mouse results in chronic elevation of serum LH and is the primary cause of the ovarian phenotype of polycystic follicles and anovulation. Prolonged treatment with a GnRH antagonist reduced serum LH levels and prevented the alphaERKO cystic ovarian phenotype. To investigate a direct role for ERalpha within the ovary, immature alphaERKO females were stimulated to ovulate with exogenous gonadotropins. Ovulatory capacity in the immature alphaERKO female was reduced compared with age-matched wild-type (14.5+/-2.9 vs. 40.6+/-2.6 oocytes/animal, respectively); however, oocytes collected from the alphaERKO were able to undergo successful in vitro fertilization. A similar discrepancy in oocyte yield was observed after superovulation of peripubertal (42 days) wild-type and alphaERKO females. In addition, ovaries from immature superovulated alphaERKO females possessed several ovulatory but unruptured follicles. Investigations of the possible reasons for the reduced number of ovulations in the alphaERKO included ribonuclease protection assays to assess the mRNA levels of several markers of follicular maturation and ovulation, including ERbeta, LH-receptor, cyclin-D2, P450-side chain cleavage enzyme, prostaglandin synthase-2, and progesterone receptor. No marked differences in the expression pattern for these mRNAs during the superovulation regimen were observed in the immature alphaERKO ovary compared with that of the wild-type. Serum progesterone levels just before ovulation were slightly lower in the alphaERKO compared with wild-type. These studies indicate that treatment of alphaERKO females with a GnRH antagonist decreased the serum LH levels to within the wild-type range and concurrently prevented development of the characteristic ovarian phenotype of cystic and hemorrhagic follicles. Furthermore, a lack of functional ERalpha within the ovary had no effect on the regulation of several genes required for follicular maturation and ovulation. However, the reduced numbers of ovulations following the administration of exogenous gonadotropins in the alphaERKO suggests an intraovarian role for ERalpha in follicular development and ovulation.


Assuntos
Ovulação , Síndrome do Ovário Policístico/genética , Receptores de Estrogênio/deficiência , Receptores de Estrogênio/genética , Animais , Gonadotropina Coriônica/farmacologia , Receptor alfa de Estrogênio , Feminino , Fertilização in vitro , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Gonadotropinas Equinas/farmacologia , Hipotálamo/fisiopatologia , Hormônio Luteinizante/sangue , Camundongos , Camundongos Knockout , Oócitos/fisiologia , Ovário/patologia , Fenótipo , Hipófise/fisiopatologia , Síndrome do Ovário Policístico/patologia , Síndrome do Ovário Policístico/fisiopatologia , Progesterona/sangue , RNA Mensageiro/metabolismo , Receptores de Estrogênio/fisiologia , Superovulação
3.
4.
Endocrinology ; 139(10): 4092-101, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9751487

RESUMO

Testicular androgens are integral components of the hormonal feedback loops that regulate circulating levels of LHbeta and FSH. The sites of feedback include hypothalamic areas regulating GnRH neurons and pituitary gonadotropes. To better define the roles of androgen receptor (AR), estrogen receptor-alpha (ERalpha), and estrogen receptor-beta (ERbeta) in mediating feedback effects of sex steroids on reproductive neuroendocrine function, we have determined the effects of castration and steroid replacement therapy on hypothalamic GnRH content, pituitary LHbeta and FSHbeta messenger RNA (mRNA) levels, and serum gonadotropins in male wild-type (WT) and estrogen receptor-alpha knockout (ERKO) mice. Hypothalami from intact WT and ERKO males contained similar amounts of GnRH, whereas castration significantly reduced GnRH contents in both genotypes. Replacement therapy with estradiol (E2), testosterone (T), or dihydrotestosterone (DHT) restored hypothalamic GnRH content in castrated (CAST) WT mice; only the androgens were effective in CAST ERKOs. Analyses of pituitary function revealed that LHbeta mRNA and serum LHbeta levels in intact ERKOs were 2-fold higher than those in intact WT males. Castration increased levels of LHbeta mRNA (1.5- to 2-fold) and serum LHbeta (4- to 5-fold) in both genotypes. Both E2 and T treatments significantly suppressed LHbeta mRNA and serum LH levels in CAST WT males. However, E2 was completely ineffective, and T was only partially effective in suppressing these two indexes in the CAST ERKO males. DHT treatments stimulated a 50% increase in LHbeta mRNA and serum LH levels in WT males, whereas serum LH was significantly suppressed in DHT-treated ERKO males. Although the pituitaries from intact ERKO males contained similar amounts of FSHbeta mRNA, serum FSH levels were 20% higher than those in the intact WT males. Castration increased FSHbeta mRNA levels only in WT males, but significantly increased serum FSH levels in both genotypes. Both E2 and T treatments significantly suppressed serum FSH in CAST WT males, whereas only E2 suppressed FSHbeta mRNA. DHT treatments of CAST WT mice stimulated a small increase in serum FSH, but failed to alter FSHbeta mRNA levels. None of the steroid treatments exerted any significant effect on FSHbeta mRNA or serum FSH levels in CAST ERKOs. These data suggest that hypothalamic GnRH contents can be maintained solely through AR signaling pathways. However, normal regulation of gonadotrope function requires aromatization of T and activation of ERalpha signaling pathways in the gonadotrope. In addition, serum FSH levels in male ERKOs appear to be regulated largely by nonsteroidal testicular factors such as inhibin. Finally, these data suggest that hypothalamic ERbeta may not be involved in mediating the negative feedback effects of T on serum LH and FSH in male mice.


Assuntos
Hormônio Foliculoestimulante/sangue , Hormônios Esteroides Gonadais/farmacologia , Hormônio Liberador de Gonadotropina/análise , Hipotálamo/química , Hormônio Luteinizante/sangue , Receptores de Estrogênio/fisiologia , Animais , Castração , Estradiol/sangue , Hormônio Foliculoestimulante/genética , Hormônio Luteinizante/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/análise
5.
Steroids ; 63(3): 149-57, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9558716

RESUMO

Diethylstilbestrol (DES) is a well-characterized carcinogen in humans and animals although its mechanisms of carcinogenicity are not yet known. While the estrogenic activity of DES is important, there is evidence that oxidative metabolism also plays an important role for its toxicity. DES is oxidatively metabolized in vivo and in vitro to a number of compounds including diethylstilbestrol-4',4"-quinone (DQ), an unstable and reactive intermediate, and Z,Z-dienestrol (ZZ-DIEN). Estrogen receptor (ER) binding assays with mouse uterine cytosol indicate that DES, DQ and ZZ-DIEN have relative binding affinities of 286, 3.6 and 0.3, respectively, relative to estradiol as 100. In addition, DQ binds irreversibly and specifically to ER suggesting that DQ may be biologically active despite its rapid metabolism and lower binding affinity compared to DES. To test this, COS-1 cells were transfected with an estrogen responsive reporter construct containing of VitA2 estrogen response element (ERE) with or without an ER expression vector. In the presence of ER, treatments with DES, DQ and ZZ-DIEN resulted in 11, 10, and 2-fold induction of chloramphenicol acetyltransferase (CAT) activity, respectively. This induction was mediated by estrogen receptor since it was suppressed by pretreatment with a 10-fold excess of the pure antiestrogen ICI 182,780. These data indicate that DQ is a biologically active intermediate that is capable of transactivation of estrogen responsive genes through the ER. Furthermore, the data suggest that the ability of DQ to irreversibly bind ER may result in persistent stimulation of ER. This persistent stimulation may be related to the carcinogenicity of DES.


Assuntos
Dietilestilbestrol/análogos & derivados , Regulação da Expressão Gênica/efeitos dos fármacos , Receptores de Estrogênio/genética , Animais , Células COS , Cloranfenicol O-Acetiltransferase/genética , Cromatografia Líquida de Alta Pressão , Dietilestilbestrol/metabolismo , Dietilestilbestrol/farmacologia , Feminino , Camundongos , Oxirredução , Receptores de Estrogênio/metabolismo , Transfecção
6.
Endocrinology ; 138(11): 4613-21, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9348186

RESUMO

Until recently, only a single type of estrogen receptor (ER) was thought to exist and mediate the genomic effects of the hormone 17beta-estradiol in mammalian tissues. However, the cloning of a gene encoding a second type of ER, termed ERbeta, from the mouse, rat, and human has prompted a reevaluation of the estrogen signaling system. Based on in vitro studies, the ERbeta protein binds estradiol with an affinity similar to that of the classical ER (now referred to as ERalpha) and is able to mediate the effects of estradiol in transfected mammalian cell lines. Essential to further investigations of the possible physiological roles of ERbeta, and its possible interactions with ERalpha, are data on the tissue distribution of the two ER types. Herein, we have described the optimization and use of an RNase protection assay able to detect and distinguish messenger RNA (mRNA) transcripts from both the ERalpha and ERbeta genes in the mouse. Because this assay is directly quantitative, a comparison of the levels of expression within various tissues was possible. In addition, the effect of disruption of the ERalpha gene on the expression of the ERbeta gene was also investigated using the ERalpha-knockout (ERKO) mouse. Transcripts encoding ERalpha were detected in all the wild-type tissues assayed from both sexes. In the female reproductive tract, the highest expression of ERbeta mRNA was observed in the ovary and showed great variation among individual animals; detectable levels were observed in the uterus and oviduct, whereas mammary tissue was negative. In the male reproductive tract, significant expression of ERbeta was seen in the prostate and epididymis, whereas the testes were negative. In other tissues of both sexes, the hypothalamus and lung were clearly positive for both ERalpha and ERbeta mRNA. The ERKO mice demonstrated slightly reduced levels of ERbeta mRNA in the ovary, prostate, and epididymis. These data, in combination with the several described phenotypes in both sexes of the ERKO mouse, suggest that the biological functions of the ERbeta protein may be dependent on the presence of ERalpha in certain cell types and tissues. Further characterization of the physiological phenotypes in the ERKO mice may elucidate possible ERbeta specific actions.


Assuntos
RNA Mensageiro/metabolismo , Receptores de Estrogênio/genética , Animais , Encéfalo/metabolismo , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Feminino , Genitália Feminina/metabolismo , Genitália Masculina/metabolismo , Masculino , Glândulas Mamárias Animais/metabolismo , Camundongos , Camundongos Knockout/genética , Hibridização de Ácido Nucleico , Hipófise/metabolismo , Ribonucleases , Distribuição Tecidual
7.
Mol Endocrinol ; 11(6): 674-81, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9171231

RESUMO

Targeted insertional disruption of the mouse estrogen receptor-alpha (ER alpha) gene has provided a genetic model in which to test hypotheses that estrogens exert important effects in development and homeostatic functions of the anterior pituitary gland, particularly in the lactotroph and gonadotroph cell types. Analysis of ER alpha gene-disrupted mice reveals a marked reduction in PRL mRNA and a decrease in lactotroph cell number, but normal specification of lactotroph cell phenotype. Gonadotropin mRNA levels in ER alpha gene-disrupted female mice are elevated, consistent with previously described transcriptional suppression of gonadotropin subunit gene expression in response to sustained administration of estrogen in wild type mice. These results provide genetic evidence that ER alpha plays a critical role in PRL and gonadotropin gene transcription and is involved in lactotroph cell growth, but is not required for specification of lactotroph cell phenotype.


Assuntos
Gonadotropinas/genética , Adeno-Hipófise/metabolismo , Prolactina/genética , Receptores de Estrogênio/fisiologia , Animais , Animais Selvagens , Feminino , Regulação da Expressão Gênica , Humanos , Técnicas Imunoenzimáticas , Masculino , Camundongos , Ovariectomia , Fenótipo , Adeno-Hipófise/citologia , Ratos , Receptores de Estrogênio/genética
8.
Trends Endocrinol Metab ; 8(4): 137-45, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-18406799

RESUMO

Disruption of the estrogen receptor (ER) gene in mice causes infertility in both sexes. Infertility in female ER knockout (ERKO) mice results from altered development of accessory sex structures, disrupted endocrine physiology, and disrupted gametogenesis. Male accessory sex structures appear relatively normal, with infertility stemming from altered sexual behaviors and disrupted gametogenesis. These findings provide significant insights into the biological importance of the ER and suggest further areas for examining the impact of estrogens on reproductive biology.

9.
Proc Natl Acad Sci U S A ; 93(22): 12626-30, 1996 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-8901633

RESUMO

Past studies have shown that epidermal growth factor (EGF) is able to mimic the uterotropic effects of estrogen in the rodent. These studies have suggested a "cross-talk" model in which EGF receptor (EGF-R) signaling results in activation of nuclear estrogen receptor (ER) and its target genes in an estrogen-independent manner. Furthermore, in vitro studies have indicated the requirement for ER in this mechanism. To verify the requirement for ER in an in vivo system, EGF effects were studied in the uteri of ER knockout (ERKO) mice, which lack functional ER. The EGF-R levels, autophosphorylation, and c-fos induction were observed at equivalent levels in both genotypes indicating that removal of ER did not disrupt the EGF responses. Induction of DNA synthesis and the progesterone receptor gene in the uterus were measured after EGF treatment of both ERKO and wild-type animals. Wild-type mice showed increases of 4.3-fold in DNA synthesis, as well as an increase in PR mRNA after EGF treatment. However, these responses were absent in ERKO mice, confirming that the estrogen-like effects of EGF in the mouse uterus do indeed require the ER. These data conclusively demonstrate the coupling of EGF and ER signaling pathways in the rodent reproductive tract.


Assuntos
Fator de Crescimento Epidérmico/farmacologia , Receptores de Estrogênio/fisiologia , Transdução de Sinais , Alelos , Animais , Sequência de Bases , Eletroforese em Gel de Poliacrilamida , Feminino , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Fosforilação , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores de Estrogênio/genética , Útero/metabolismo
10.
J Clin Endocrinol Metab ; 81(6): 2055-60, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8964828

RESUMO

Conversion of testosterone to dihydrotestosterone (DHT) has been demonstrated to be catalyzed by two isoforms of steroid 5 alpha-reductase, designated types I and II. Although several classes of steroid-based inhibitors of the type II isoform have been identified, these agents have not demonstrated highly selective pharmacological activity against human type I 5 alpha-reductase. LY191704 is representative of a series of nonsteroidal agents that have potent [apparent inhibitory constant (Ki) = 11.3 nM] inhibitory activity in human scalp skin homogenates (pH 7.5), a source of type I 5 alpha-reductase. [3H]-DHT production in the presence and absence of LY191704 is consistent with a noncompetitive mode of inhibition. In human prostatic homogenates (pH 5.5), a source of type II 5 alpha-reductase, LY191704 is virtually inactive as an inhibitor [concentration of inhibitor producing 50% inhibition of enzymatic activity (IC50) > 1,000 nM] of [3H]-DHT formation. LY191704 does not inhibit the type I or type II isoforms of rat 5 alpha-reductase, nor does the compound compete for binding to the murine androgen receptor expressed in SF9 cells using a baculo virus expression system. The benzoquinolinones, as exemplified by LY191704, possess exquisite pharmacological selectivity and provide a tool to understand the role of human type I 5 alpha-reductase in normal and pathophysiological states. These agents may also find clinical utility in treating androgen-dependent dermatological conditions.


Assuntos
Inibidores de 5-alfa Redutase , Isoenzimas/antagonistas & inibidores , Quinolonas/farmacologia , Couro Cabeludo/enzimologia , Animais , Ligação Competitiva , Di-Hidrotestosterona/metabolismo , Humanos , Masculino , Camundongos , Concentração Osmolar , Quinolonas/metabolismo
11.
Recent Prog Horm Res ; 51: 159-86; discussion 186-8, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8701078

RESUMO

The estrogen receptor (ER) is thought to play a crucial role in the regulation of many life processes, including development, reproduction and normal physiology. Because there have been no known mutations of the estrogen receptor in normal tissue of humans and animals, its presence and tissue distribution is thought to be essential for survival. Using the techniques of homologous recombination, we have disrupted the ER gene and have produced a line of transgenic mice possessing the altered ER gene (ERKO). The mouse ER gene was disrupted by inserting a 1.8 kb PGK-Neomycin sequence into exon 2, approximately 280 bp downstream of the transcription start codon. The correct targeting of the disruption was demonstrated by Southern blot analysis and PCR. Western blot analysis of uterine preparations from ERKO females showed no detectable ER protein. Heterozygotes had one half the level of ER protein compared to wild-type animals. Estrogen insensitivity was confirmed using estrogen agonists, estradiol, hydroxy tamoxifen, diethylstilbestrol treatment for 3 days which resulted in a 3-4-fold increase in uterine wet weight and vaginal cornification in wild-type females, while ERKO mice were totally unresponsive. These data were further supported by the failure of estrogen or EGF treatment to induce DNA synthesis in uterine tissue of similarly treated mice. Lactoferrin, an estrogen-responsive gene in the uterus, was also assayed by Northern blot. Wild-type mice treated with a single estradiol injection showed a 350-fold induction in lactoferrin mRNA. while ERKO females showed no detectable response. Both male and female animals survive to adulthood with normal gross external phenotypes. As expected, females are infertile and demonstrate hypoplastic uteri and hyperemic ovaries with no apparent corpora lutea. Males are also infertile, with atrophy of the testes and seminiferous tubule dysmorphogenesis. Although the reproductive capabilities have been altered with a dramatic effect on the gonads, prenatal development of the reproductive tracts of both sexes appear to be independent of an ER-mediated response. Analysis of the mammary glands of the ERKO females at 4 months of age showed a primitive ductal rudiment rather than the fully developed ductal tree seen in wild-type siblings. Also absent were the terminal end buds seen during normal ductal morphogenesis. Both sexes show a decrease in skeletal bone density, supporting a direct role for ER action in bone. A single patient is described who is homozygous for a point mutation in the human ER gene at codon 157. The mutation produces a truncation of the ER protein and results in estrogen insensitivity syndrome. Most significant of the clinical findings are effects on skeletal bone density and retarded bone age. Findings from the patient and mice suggest that the absence of functional ER is not lethal. Mutation in the ER gene is present in the human population. Further characterization of the mice and identification of additional patients will be required to more fully understand the consequences of ER gene mutations.


Assuntos
Mutação , Fenótipo , Receptores de Estrogênio/genética , Receptores de Estrogênio/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Estrogênios/farmacologia , Estrogênios/fisiologia , Feminino , Marcação de Genes , Humanos , Masculino , Camundongos , Dados de Sequência Molecular , Linhagem , Receptores de Estrogênio/química
12.
Mol Endocrinol ; 9(11): 1441-54, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8584021

RESUMO

We employed homologous recombination in mouse embryonic stem cells to disrupt the estrogen receptor (ER) gene. Subsequently generated mice that are homozygous for the gene disruption, termed ERKO, possess no demonstrable wild-type ER by Western blot analysis. However, the presence of residual high affinity binding, as detected by [3H]estradiol binding assays and sucrose gradients in uterine extracts from ERKO females prompted further investigation of transcription and translation products from the disrupted ER gene. Analysis of ERKO uterine messenger RNA (mRNA) by reverse transcriptase-polymerase chain reaction demonstrated that although no full-length wild-type ER mRNA was present, two smaller transcripts, labeled E1 and E2, were identified and partially sequenced. Both ERKO transcripts are splicing variants that result in the disrupting NEO sequence being partially or completely removed from the mRNA. In the ERKO-E2 variant, this results in a frame shift and the creation of at least two stop codons downstream. In the ERKO-E1 variant, the ER reading frame is preserved and encodes for a smaller mutant ER that could be the source of the residual estradiol binding. When this mutant form is overexpressed and characterized in vitro, it results in a smaller protein of the predicted size that possesses significantly reduced estrogen-dependent transcriptional activity compared with that of the wild-type ER. Despite residual amounts of an impaired ER variant, estrogen insensitivity in the female ERKOs was confirmed by the failure of estrogen treatment to induce known uterine markers of estrogen action, such as increased DNA synthesis, and transcription of the progesterone receptor, lactoferrin, and glucose-6-phosphate dehydrogenase genes. Furthermore, serum levels of estradiol in the ERKO female are more than 10-fold higher than those in the wild type, consistent with a syndrome of hormone insensitivity.


Assuntos
Estradiol/farmacologia , Marcação de Genes , Receptores de Estrogênio/genética , Transcrição Gênica/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Linhagem Celular Transformada , Chlorocebus aethiops , Resistência a Medicamentos/genética , Estradiol/sangue , Feminino , Mutação da Fase de Leitura , Regulação da Expressão Gênica/efeitos dos fármacos , Glucosefosfato Desidrogenase/biossíntese , Glucosefosfato Desidrogenase/genética , Lactoferrina/biossíntese , Lactoferrina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Ovariectomia , Progesterona/sangue , Biossíntese de Proteínas/efeitos dos fármacos , Splicing de RNA , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores de Estrogênio/biossíntese , Receptores de Estrogênio/deficiência , Receptores de Estrogênio/fisiologia , Receptores de Progesterona/biossíntese , Receptores de Progesterona/genética , Células-Tronco , Transfecção , Útero/efeitos dos fármacos , Útero/metabolismo
13.
Endocrinology ; 136(5): 2172-8, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7720667

RESUMO

Elevation of intracellular calcium levels in the presence of normal androgen levels has been implicated in apoptotic prostate cell death. Since the androgen receptor (AR) plays a critical role in the regulation of growth and differentiation of the prostate, it was of interest to determine whether Ca2+ would affect the expression of androgen receptor messenger RNA (mRNA) and protein, thus affecting the ability of androgens to control prostate function. AR-positive human prostate cancer cells, LNCaP, were incubated with either the calcium ionophore A23187 or the intracellular endoplasmic reticulum Ca(2+)-ATPase inhibitor thapsigargin. Subsequently, AR mRNA and protein levels were assessed by Northern and Western blot analysis. Both A23187 and thapsigargin were found to down-regulate steady state AR mRNA levels in a time- and dose-dependent manner. AR mRNA began to decrease after 6-8 h of incubation with 10(-6) M A23187 or 10(-7) M thapsigargin, reaching a nadir at 16 and 10 h of incubation, respectively. In contrast, control mRNA (glyceraldehyde 3-phosphate dehydrogenase) did not change significantly during the treatments with either A23187 or thapsigargin. AR protein levels were found to be decreased after 12 h of incubation with either 10(-6) M A23187 or 10(-7) M thapsigargin. The decrease in AR mRNA and protein seemed to precede apoptosis, since neither A23187 (24 h) nor thapsigargin (30 h) was found to alter cell morphology within the treatment time. Cycloheximide and actinomycin D were unable to change the calcium-mediated decrease in AR mRNA, ruling out the necessity for de novo protein synthesis or a change in mRNA stability. Moreover, the decrease in AR mRNA induced by calcium does not seem to involve protein kinase C- or calmodulin-dependent pathways, since inhibitors of these cellular components had no effect. Nuclear run-on assays demonstrated little or no effects of either A23187 or thapsigargin treatment on AR gene transcription (8 h and 10 h). In conclusion, these studies show that intracellular calcium seems to be a potent regulator of AR gene expression in LNCaP cells.


Assuntos
Calcimicina/farmacologia , Cálcio/metabolismo , Expressão Gênica , Receptores Androgênicos/biossíntese , Terpenos/farmacologia , Alcaloides/farmacologia , Apoptose/efeitos dos fármacos , Northern Blotting , Western Blotting , ATPases Transportadoras de Cálcio/antagonistas & inibidores , Linhagem Celular , Núcleo Celular/metabolismo , Dactinomicina/farmacologia , Relação Dose-Resposta a Droga , Expressão Gênica/efeitos dos fármacos , Humanos , Cinética , Masculino , Neoplasias da Próstata , Proteína Quinase C/antagonistas & inibidores , RNA Mensageiro/biossíntese , RNA Mensageiro/isolamento & purificação , RNA Neoplásico/biossíntese , RNA Neoplásico/isolamento & purificação , Estaurosporina , Tapsigargina , Células Tumorais Cultivadas
14.
Biochemistry ; 33(48): 14594-600, 1994 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-7981221

RESUMO

The androgen receptor (AR) is a developmental and tissue-specific transcription factor which is activated by binding testosterone or dihydrotestosterone. Several different methods of transcriptional regulation of the AR have been shown, including regulation by androgens, follicle-stimulating hormone, epidermal growth factor, and the cAMP pathway. In order to further characterize the transcriptional regulation of the AR, portions of the mouse androgen receptor (mAR) promoter were cloned into the promoterless pBLCAT3 vector and assayed for chloramphenicol acetyltransferase activity. The results indicate that in addition to the previously characterized promoter (+1) there is a second distinct promoter located 3' to the first promoter. Amplification of the 5'-end of the AR gene indicates that RNA originating from the second promoter is initiated from 162 and 170 bases downstream from the 5'-most previously characterized site. Northern blot analysis indicated that RNA initiated from the two promoters is differentially expressed in several cell lines and multiple tissues. Androgen ablation by castration showed that both promoters are controlled by androgens in the kidney. Sequence analysis revealed that the second promoter does not contain a TATA or CAAT box. Further characterization of this promoter may provide important insights into the transcriptional regulation of the androgen receptor since previous studies have often included only the first promoter.


Assuntos
Di-Hidrotestosterona/farmacologia , Regiões Promotoras Genéticas , Receptores Androgênicos/genética , Animais , Primers do DNA/química , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Orquiectomia , RNA Mensageiro/genética , Transcrição Gênica/efeitos dos fármacos , Transfecção
15.
Prostate ; 25(1): 39-45, 1994 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8022710

RESUMO

Recent studies have demonstrated that retinoic acid (RA) can repress the growth of human prostatic epithelial cells. Since the proliferation of prostate cells is highly dependent on androgen stimulation, presumably via its cognate receptor, we investigated the effects of RA on the expression of the androgen receptor and other androgen-regulated genes in the human prostatic adenocarcinoma cell line LNCaP. Using a radioligand binding assay, we found that androgen-binding activity was reduced 30-40% in cells treated with 10(-5) M RA plus 6 nM dihydrotestosterone (DHT), as compared to cells with the androgen alone. Moreover, the reduction of the androgen receptor (AR) was not accompanied by alteration of the ligand-binding affinity. Concomitant changes in the function of AR were manifested by a dramatic reduction in AR-mediated transcription activity in a transfection experiment. Androgen-induced levels of both prostate-specific antigen (PSA) and human glandular kallikrein-1 (hKLK2) mRNAs were significantly repressed by RA in a dose- and time-dependent manner. Consistent with this finding, androgen induction of PSA glycoprotein was also repressed by RA, with maximal inhibition occurring at 10(-5) M. These data suggest that the suppression of proliferation and function of prostatic cells by RA may be via modulatory effects on the AR.


Assuntos
Antagonistas de Androgênios/farmacologia , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/efeitos dos fármacos , Tretinoína/farmacologia , Adenocarcinoma/metabolismo , Divisão Celular/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Hibridização de Ácido Nucleico , Neoplasias da Próstata/patologia , RNA Neoplásico/efeitos dos fármacos , Ensaio Radioligante , Receptores Androgênicos/análise , Células Tumorais Cultivadas
16.
Mol Endocrinol ; 8(4): 448-55, 1994 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8052266

RESUMO

The androgen receptor (AR) mediates the biological functions of androgens and is essential for normal growth and differentiation of urogenital organs as well as initiation and maintenance of spermatogenesis. Withdrawal of androgens by castration or other methods has been shown to cause a marked, although often temporary, regression of many prostate cancers. In order to gain a better understanding of the transcriptional regulation of the AR, a series of truncation mutants derived from the 5'-region of the mouse AR (mAR) were inserted into the promoter-less plasmid pBLCAT3 and transiently expressed in the mouse alpha T3-1 and GT1-7 cell lines. The results of these experiments indicate the presence of a negative regulatory element in the 5'-untranslated region of the gene, which is able to reduce chloramphenicol acetyltransferase (CAT) activity by 77-89%. We have named this element the mAR suppressor (mARS). DNase-I protection assays of the 5'-untranslated region disclosed a protected domain. Gel mobility assays using the mARS revealed the presence of three protein-DNA complexes that could specifically bind to this protected domain. Insertion of the mARS into the thymidine kinase promoter containing pBLCAT2 vector resulted in a 2- to 10-fold decrease in CAT activity, but only if the insert was 3' to the start of transcription initiation. Finally, point mutations within the mARS were able to increase transcription of the AR promoter by 2.3-fold. The results of these experiments indicate that the mAR 5'-untranslated region contains a suppressor element.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Regulação da Expressão Gênica , Genes , Camundongos/metabolismo , Regiões Promotoras Genéticas , Receptores Androgênicos/genética , Sequências Reguladoras de Ácido Nucleico , Transcrição Gênica , Animais , Sequência de Bases , Linhagem Celular , Núcleo Celular/metabolismo , Cloranfenicol O-Acetiltransferase/biossíntese , Cloranfenicol O-Acetiltransferase/genética , Sequência Consenso , Hipotálamo/citologia , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Hipófise/citologia , Mutação Puntual , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Deleção de Sequência , Simplexvirus/genética , Timidina Quinase/genética
17.
Vitam Horm ; 49: 383-432, 1994.
Artigo em Inglês | MEDLINE | ID: mdl-7810074

RESUMO

Androgens directly regulate a vast number of physiological events. These direct androgen effects are mediated by a nuclear receptor that exhibits four major functions or activities: steroid binding, DNA binding, transactivation, and nuclear localization. The SBD consists of a hydrophobic pocket of amino acids that exhibits high-affinity, androgen-specific binding. Based on studies of mutant AR, it appears that a number of different amino acids contribute to the steroid binding characteristics of the AR. The DNA binding domain confers sequence-specific binding to structures called androgen-responsive elements. The specificity of steroid binding and DNA binding provides a crucial basis for androgen-specific regulation of target genes. The nuclear localization signal shares homology with known nuclear localization signals and, coupled with the presence of androgens, is responsible for localizing the AR to the nucleus. The transactivation functions reside mostly in the NH2 terminus but the responsible domains are as yet poorly defined. Though the different domains can act as independent moieties, one domain can clearly alter the behavior of another domain. For instance, the SBD appears to inhibit the transactivating functions until steroid is bound and the amino terminus prevents DNA binding activity until steroid is bound. The relative ease of introducing mutations with polymerase chain reaction technology will facilitate further delineation of critical amino acids and domains responsible for the various activities of the AR. The recent cloning and characterization of AR promoters revealed that the AR genes are driven by a TATA-less promoter characteristics of housekeeping genes. Analysis of transcription rates, mRNA levels, and protein levels indicates that androgens and pkA and pkC pathways modulate expression of AR mRNA and protein. This indicates that the same signal pathways that interact to regulate androgen target genes also regulate the levels of AR in the target tissues. Surprisingly few androgen-regulated genes have been well characterized for the mechanisms by which androgen regulates the gene. The C(3), Slp, probasin, PSA, and hKLK2 genes have provided examples where androgens regulate transcription. Posttranscriptional regulation by androgens has been demonstrated for the SVP1, 2, 3, and 4 and AR genes. The mechanisms underlying posttranscriptional regulation are poorly defined but substantial progress has been made in defining the critical elements that mediate transcriptional effects of androgens. Transcriptional effects are mediated through binding of androgen-AR complexes to specific DNA sequences called AREs. Simple AREs such as those found in C(3) and kallikrein genes tend to be permissive in that GR and PR can also act through the same element.(ABSTRACT TRUNCATED AT 400 WORDS)


Assuntos
Androgênios/fisiologia , Receptores Androgênicos/fisiologia , Androgênios/genética , Animais , Sequência de Bases , Regulação da Expressão Gênica , Humanos , Dados de Sequência Molecular , Oligodesoxirribonucleotídeos , Receptores Androgênicos/química , Relação Estrutura-Atividade
18.
Receptor ; 4(2): 121-34, 1994.
Artigo em Inglês | MEDLINE | ID: mdl-7950980

RESUMO

Androgen insensitivity in the testicular feminized (Tfm) mouse is caused by frame-shift mutation in the androgen receptor (AR) mRNA, which results in a stop codon in the amino terminus. Despite this mutation, a smaller sized protein corresponding to the DNA- and steroid-binding domain of the AR can be synthesized from the cloned Tfm AR cDNA by in vitro translation. The Tfm AR construct was demonstrated to express a protein capable of binding androgen with an affinity similar to the cloned wild-type AR. Although the Tfm AR product failed to transactivate mouse mammary tumor virus-long terminal repeat (MMTV-LTR) promoter when low concentrations (100 ng) of Tfm AR vector were cotransfected, higher concentrations (5000 ng) resulted in a residual amount of transactivation, suggesting lower level transactivating capabilities. By cotransfecting the Tfm AR expression vector with the wild-type receptor, it was demonstrated that the product of the Tfm AR gene is capable of inhibiting the transactivation activity of the wild-type receptor. These data suggest that although the Tfm AR mRNA fails to produce a full-length AR because of the frame-shift mutation, a smaller protein capable of binding both steroid and DNA can be produced by translation from an internal initiation codon. This product could account for the low levels of androgen-binding activity detected previously in the Tfm mouse.


Assuntos
Síndrome de Resistência a Andrógenos/genética , Androgênios/metabolismo , Iniciação Traducional da Cadeia Peptídica , Receptores Androgênicos/genética , Animais , Sequência de Bases , Mutação da Fase de Leitura , Masculino , Camundongos , Camundongos Mutantes , Dados de Sequência Molecular , Receptores Androgênicos/biossíntese , Síndrome , Ativação Transcricional
19.
Mol Endocrinol ; 7(12): 1530-40, 1993 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-7511785

RESUMO

Both androgens and cAMP-mediated hormones are known to regulate expression of the androgen receptor (AR) gene. In order to determine whether these effects occur at the transcriptional level, transfection studies were conducted with a 1.5-kilobase fragment of the 5'-flanking region of the mouse AR gene coupled to a chloramphenicol acetyltransferase (CAT) reporter gene. We demonstrated that the cAMP pathway regulates expression of the mouse AR (mAR) 5'-CAT construct in mouse pituitary (alpha T3-1), rat pituitary (GC), and quail fibroblast (QT6) cell lines. Deletional analysis indicated that several areas of this clone, including a region containing a putative cAMP response element (CRE), are involved in mediating cAMP regulation of the AR gene. Oligonucleotides containing a putative CRE, linked to the thymidine kinase promoter of pBLCAT2, conferred increased forskolin induction of CAT activity. Furthermore, overexpression of a CRE binding protein up-regulated expression of the mAR 5'-CAT constructs. Bandshift data demonstrated that the putative CRE forms specific, competable complexes with nuclear proteins from alpha T3-1 and QT6 cells. Additional experiments indicated that AR can modulate both basal and forskolin-induced CAT activity in an androgen-dependent fashion. These data provide evidence that the 5'-flanking region of the mAR gene contains sequences which mediate the effects of both cAMP and androgens.


Assuntos
Androgênios/fisiologia , AMP Cíclico/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Receptores Androgênicos/genética , 1-Metil-3-Isobutilxantina/farmacologia , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Animais , Sequência de Bases , Células Cultivadas , Colforsina/farmacologia , Coturnix , Di-Hidrotestosterona/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Genes , Camundongos , Dados de Sequência Molecular , Adeno-Hipófise/efeitos dos fármacos , Adeno-Hipófise/metabolismo , Regiões Promotoras Genéticas , Receptores Androgênicos/biossíntese , Proteínas Recombinantes de Fusão/biossíntese , Sequências Reguladoras de Ácido Nucleico , Deleção de Sequência
20.
Horm Behav ; 27(2): 269-81, 1993 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8349284

RESUMO

Progestins often act as potent antiandrogens in male birds and mammals. Experiments with lizards find that progestins can both inhibit (when given in high dosages) or stimulate (when given in low dosages) male-typical sex behavior in gonadectomized individuals. This study shows that in the little striped whiptail lizard exogenous progesterone (P) facilitates androgen-dependent sex behaviors in males yet fails to stimulate seasonal activation of androgen-dependent accessory sex structures. Analysis of androgen receptors (AR) in brain and kidney cytosol of the little striped whiptail lizard reveals similarities with the AR of the mouse. The data indicate that despite the ability of P to mimic the actions of androgens in activating sex behaviors in males of this species, the characteristics of the AR are conserved with respect to other vertebrate species.


Assuntos
Di-Hidrotestosterona/farmacologia , Lagartos/fisiologia , Progesterona/farmacologia , Receptores Androgênicos/efeitos dos fármacos , Comportamento Sexual Animal/efeitos dos fármacos , Maturidade Sexual/efeitos dos fármacos , Animais , Copulação/efeitos dos fármacos , Copulação/fisiologia , Citosol/efeitos dos fármacos , Citosol/fisiologia , Feminino , Rim/anatomia & histologia , Rim/efeitos dos fármacos , Masculino , Orquiectomia , Receptores Androgênicos/fisiologia , Comportamento Sexual Animal/fisiologia , Maturidade Sexual/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...