Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
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.

2.
Int J Dev Neurosci ; 78: 33-44, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31400491

RESUMO

To further reveal the molecular mechanism underlying sexual differentiation of the mouse cerebral cortex and hippocampus, we reanalyzed our previous microarray study with Gene Ontology (GO) term enrichment and found that the GO term "RNA binding" was over-represented among the 89 sexually dimorphic candidate genes. Thus, we selected 16 autosomal genes annotated to the term RNA binding and profiled their mRNA expression in the developing male and female mouse cortex/hippocampus. During the first three weeks after birth, sex differences in mRNA levels of Khdrbs2, Nanos2, Rbm48, and Tdrd3 were observed in the mouse cortex/hippocampus. Of these genes, only the female-biased expression of Rbm48 in neonates was abolished by prenatal exposure to testosterone propionate (TP), while postnatal treatment of TP three weeks after birth increased Rbm48 and Tdrd3 mRNA levels in both sexes. Regardless of sex, the postnatal cortex/hippocampus also showed a marked increase in the content of androgen receptor (Ar) and estrogen receptor ß (Esr2), but a decrease in estrogen receptor α (Esr1) and aromatase (Cyp19a1), which might confer the different responses of Rbm48 to prenatal and postnatal TP. Our results suggest that androgen-regulated, sexually dimorphic Rbm48 expression might present a novel molecular mechanism by which perinatal androgens control development of sexual dimorphism in cortical and hippocampal structure and function.


Assuntos
Androgênios/farmacologia , Córtex Cerebral/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Proteínas de Ligação a RNA/metabolismo , Propionato de Testosterona/farmacologia , Animais , Aromatase/metabolismo , Córtex Cerebral/metabolismo , Feminino , Hipocampo/metabolismo , Masculino , Camundongos , Proteínas/genética , Proteínas/metabolismo , Proteínas de Ligação a RNA/genética , Receptores Androgênicos/metabolismo , Receptores de Estrogênio/metabolismo , Caracteres Sexuais , Fatores Sexuais
3.
Neurosci J ; 2015: 525369, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26317111

RESUMO

During the perinatal period, male mice are exposed to higher levels of testosterone (T) than females, which promotes sexual dimorphism in their brain structures and behaviors. In addition to acting via estrogen receptors after being locally converted into estradiol by aromatase, T also acts directly through androgen receptor (AR) in the brain. Therefore, we hypothesized that AR expression in the developing mouse cortex and hippocampus was sexually dimorphic. To test our hypothesis, we measured and determined AR mRNA and protein levels in mouse cortex/hippocampus collected on the day of birth (PN0) and 7 (PN7), 14 (PN14), and 21 (PN21) days after birth. We demonstrated that, as age advanced, AR mRNA levels increased in the cortex/hippocampus of both sexes but showed no sex difference. Two AR proteins, the full-length (110 kDa) and a smaller isoform (70 kDa), were detected in the developing mouse cortex/hippocampus with an age-dependent increase in protein levels of both AR isoforms at PN21 and a transient masculine increase in expression of the full-length AR protein on PN7. Thus, we conclude that the postnatal age and sex differences in AR protein expression in combination with the sex differences in circulating T may cause sexual differentiation of the mouse cortex/hippocampus.

4.
J Steroids Horm Sci ; 5(3): 1000139, 2014 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-25411648

RESUMO

OBJECTIVE: Using gene expression microarrays and reverse transcription with quantitative polymerase chain reaction (RT-qPCR), we have recently identified several novel genes that are differentially expressed in the neonatal male versus female mouse cortex/hippocampus (Armoskus et al.). Since perinatal testosterone (T) secreted by the developing testes masculinizes cortical and hippocampal structures and the behaviors regulated by these brain regions, we hypothesized that sexually dimorphic expression of specific selected genes in these areas might be regulated by T during early development. METHODS: To test our hypothesis, we treated timed pregnant female mice daily with vehicle or testosterone propionate (TP) starting on embryonic day 16 until the day of birth. The cortex/hippocampus was collected from vehicle- and TP-treated, male and female neonatal pups. Total RNA was extracted from these brain tissues, followed by RT-qPCR to measure relative mRNA levels of seven sex chromosome genes and three autosomal genes that have previously showed sex differences. RESULTS: The effect of prenatal TP was confirmed as it stimulated Dhcr24 expression in the neonatal mouse cortex/hippocampus and increased the anogenital distance in females. We found a significant effect of sex, but not TP, on expression of three Y-linked (Ddx3y, Eif2s3y, and Kdm5d), four X-linked (Eif2s3x, Kdm6a, Mid1, and Xist), and one autosomal (Klk8) genes in the neonatal mouse cortex/hippocampus. CONCLUSION: Although most of the selected genes are not directly regulated by prenatal T, their sexually dimorphic expression might play an important role in the control of sexually differentiated cognitive and social behaviors as well as in the etiology of sex-biased neurological disorders and mental illnesses.

5.
Brain Res ; 1562: 23-38, 2014 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-24661915

RESUMO

The cerebral cortex and hippocampus are important for the control of cognitive functions and social behaviors, many of which are sexually dimorphic and tightly regulated by gonadal steroid hormones via activation of their respective nuclear receptors. As different levels of sex steroid hormones are present between the sexes during early development and their receptors act as transcription factors to regulate gene expression, we hypothesize that sexually dimorphic gene expression in the developing mouse cortex and hippocampus might result in sex differences in brain structures and neural circuits governing distinct behaviors between the sexes as adults. To test our hypothesis, we used gene expression microarrays to identify 90 candidate genes differentially expressed in the neonatal cortex/hippocampus between male and female mice, including 55 male-biased and 35 female-biased genes. Among these genes, sexually dimorphic expression of eight sex chromosome genes was confirmed by reverse transcription with quantitative PCR (RT-qPCR), including three located on the X chromosome (Xist, Eif2s3x, and Kdm6a), three on the Y chromosome (Ddx3y, Eif2s3y, and Kdm5d), and two in the pseudoautosomal region of the X and Y chromosomes (Erdr1 and Mid1). In addition, five autosomal genes (Cd151, Dab2, Klk8, Meg3, and Prkdc) were also validated for their sexually dimorphic expression in the neonatal mouse cortex/hippocampus. Gene Ontology annotation analysis suggests that many of these sexually dimorphic genes are involved in histone modifications, cell proliferation/death, androgen/estrogen signaling pathways, and synaptic organization, and these biological processes have been implicated in differential neural development, cognitive function, and neurological diseases between the sexes.


Assuntos
Córtex Cerebral/metabolismo , Expressão Gênica , Hipocampo/metabolismo , Caracteres Sexuais , Animais , Animais Recém-Nascidos , Córtex Cerebral/crescimento & desenvolvimento , Análise por Conglomerados , Feminino , Perfilação da Expressão Gênica , Genes Ligados ao Cromossomo X/fisiologia , Genes Ligados ao Cromossomo Y/fisiologia , Hipocampo/crescimento & desenvolvimento , Masculino , Camundongos Endogâmicos C57BL , Análise em Microsséries , Reação em Cadeia da Polimerase em Tempo Real
6.
Behav Brain Res ; 258: 112-8, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24144547

RESUMO

The current study was to examine the relationship between the number of gonadotropin-releasing hormone (GnRH) neurons and male sexual behavior in middle-aged rats. Based on their sexual performance, middle-aged male rats (18-19 months) were assigned to three groups: (i) Group MIE (showing mounts, intromissions, and ejaculation), (ii) Group MI (displaying mounts and intromissions, but no ejaculation), and (iii) Group NC (showing no copulatory behavior). The brains of these middle-aged animals and of sexually active, young controls were collected and then examined for immunohistochemical localization of GnRH neurons. The numbers of two subtypes of GnRH neurons, smooth (s-GnRH) and irregular (i-GnRH), in the medial septum (MS), organum vasculosum of the lamina terminalis (OVLT), preoptic area (POA), and anterior hypothalamus (AH), were determined under a light microscope. As compared to young controls, an age-related decrease in the number of s-GnRH neurons was found in the MS of MIE rats. Among three groups of middle-aged rats, Groups MIE and MI had more s-GnRH neurons in the POA and i-GnRH neurons in the OVLT and POA than Group NC. In addition, loss of s-GnRH and i-GnRH neurons in the MS was observed in Groups MI and NC and Group NC, respectively. Our results suggest that a decrease in GnRH neuron subtypes occurring in different brain regions might be critical for the loss of specific components of male rat sexual behavior during aging.


Assuntos
Envelhecimento/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Septo do Cérebro/metabolismo , Comportamento Sexual Animal/fisiologia , Animais , Contagem de Células , Masculino , Ratos , Ratos Long-Evans
7.
J Neurosci ; 30(30): 9967-72, 2010 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-20668181

RESUMO

Sexual behavior is variable between individuals, ranging from celibacy to sexual addictions. Within normal populations of individual men, ranging from young to middle aged, testosterone levels do not correlate with libido. To study the genetic mechanisms that contribute to individual differences in male sexual behavior, we used hybrid B6D2F1 male mice, which are a cross between two common inbred strains (C57BL/6J and DBA/2J). Unlike most laboratory rodent species in which male sexual behavior is highly dependent upon gonadal steroids, sexual behavior in a large proportion of these hybrid male mice after castration is independent of gonadal steroid hormones and their receptors; thus, we have the ability to discover novel genes involved in this behavior. Gene expression arrays, validation of gene candidates, and transgenic mice that overexpress one of the genes of interest were used to reveal genes involved in maintenance of male sexual behavior. Several genes related to neuroprotection and neurodegeneration were differentially expressed in the hypothalamus of males that continued to mate after castration. Male mice overexpressing the human form of one of these candidate genes, amyloid beta precursor protein (APP), displayed enhanced sexual behavior before castration and maintained sexual activity for a longer duration after castration compared with controls. Our results reveal a novel and unexpected relationship between APP and male sexual behavior. We speculate that declining APP during normal aging in males may contribute to the loss of sexual function.


Assuntos
Precursor de Proteína beta-Amiloide/fisiologia , Regulação da Expressão Gênica/fisiologia , Comportamento Sexual Animal/fisiologia , Precursor de Proteína beta-Amiloide/genética , Análise de Variância , Animais , Comportamento Animal , Perfilação da Expressão Gênica/métodos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Orquiectomia/métodos , Área Pré-Óptica/metabolismo , RNA Mensageiro/metabolismo , Núcleos Septais/metabolismo , Transdução de Sinais/fisiologia
8.
Epigenetics ; 4(1): 47-53, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19029819

RESUMO

Sex differences in neural development are established via a number of cellular processes (i.e., migration, death and survival). One critical factor identified is the neonatal rise in testosterone (T) which activates gene transcription via androgen (AR) and, after aromatization to estradiol, estrogen receptors (ERalpha and beta). Recent evidence shows that AR and ERs interact with histone modifying enzymes. Post-translational modifications of histones, including acetylation and methylation, are involved in transcriptional regulation during normal development. Therefore, we hypothesized that acetylation and/or methylation of histone H3 may underlie sexual differentiation, at least in some regions of the brain. We measured levels of acetylated (H3K9/14Ac) and trimethylated (H3K9Me3) H3 in whole neonatal mouse brains and in three regions: preoptic area + hypothalamus, amygdala and cortex + hippocampus (CTX/HIP). Sex differences in H3K9/14Ac and H3K9Me3 (males > females) were noted in the CTX/HIP on embryonic day 18, the day of birth, and six days later. To determine if T mediates these changes in H3 modifications, pregnant dams received vehicle or T for the final four days of gestation; pup brains were collected at birth. Methylation of H3 was sexually dimorphic despite hormone treatment. In contrast, H3 acetylation in the CTX/HIP of females from T-treated dams rose to levels equivalent to males. Thus, H3 modifications are sexually dimorphic in the developing mouse CTX/HIP and acetylation, but not methylation, is masculinized in females by T in utero. This is the first demonstration that histone modification is associated with neural sexual differentiation.


Assuntos
Encéfalo/crescimento & desenvolvimento , Encéfalo/fisiologia , Epigênese Genética , Histonas/metabolismo , Fatores Sexuais , Acetilação , Animais , Encéfalo/metabolismo , Feminino , Regulação da Expressão Gênica , Masculino , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional , Fatores de Tempo , Ativação Transcricional
9.
J Gerontol A Biol Sci Med Sci ; 63(7): 678-82, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18693221

RESUMO

The correlation between dopamine (DA) and norepinephrine (NE) levels in the bed nucleus of the stria terminalis (BNST) and male sexual behavior was examined in middle-aged rats. Male rats (18-19 months) were divided into: (a) Group MIE, consisting of rats showing mounts, intromissions, and ejaculations; (b) Group MI, composed of rats showing mounts and intromissions, but no ejaculation; and (c) Group NC, consisting of noncopulators. Young adult rats (4-5 months) displaying complete copulatory behavior were used as the control. Tissue levels of DA, NE, and DA metabolites in the BNST were measured by high-pressure liquid chromatography. DA, but not NE, levels in MIE rats were significantly lower than those in young controls. DA and NE levels in MIE rats were significantly higher than those in NC rats. These results suggest that DA and NE in the BNST might play an important role in the control of male sexual behavior in middle-aged rats.


Assuntos
Dopamina/metabolismo , Norepinefrina/metabolismo , Núcleos Septais/metabolismo , Comportamento Sexual Animal/fisiologia , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Fatores Etários , Animais , Cromatografia Líquida de Alta Pressão , Ácido Homovanílico/metabolismo , Masculino , Ratos , Ratos Long-Evans
10.
Horm Behav ; 53(2): 342-50, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18191134

RESUMO

Neuropeptide Y (NPY) stimulates feeding, depresses sexual behavior, and its expression in the brain is modulated by energetic status. We examined the role of NPY in female musk shrews, a species with high energetic and reproductive demands; they store little fat, and small changes in energy can rapidly diminish or enhance sexual receptivity. Intracerebroventricular infusion of NPY enhanced acute food intake in shrews; however, NPY had little affect on sexual receptivity. The distribution of NPY immunoreactivity in the female musk shrew brain was unremarkable, but energy status differentially affected NPY immunoreactivity in several regions. Similar to what has been noted in other species, NPY immunoreactivity was less dense in brains of ad libitum shrews and greater in shrews subjected to food restriction. In two midbrain regions, both of which contain high levels of gonadotropin releasing hormone II (GnRH II), which has anorexigenic actions in shrews, NPY immunoreactivity was more sensitive to changes in food intake. In these regions, acute re-feeding (90-180 min) after food restriction reduced NPY immunoreactivity to levels noted in ad libitum shrews. We hypothesize that interactions between NPY and GnRH II maintain energy homeostasis and reproduction in the musk shrew.


Assuntos
Regulação do Apetite/fisiologia , Encéfalo/metabolismo , Metabolismo Energético/fisiologia , Neuropeptídeo Y/fisiologia , Comportamento Sexual Animal/fisiologia , Musaranhos/metabolismo , Animais , Relação Dose-Resposta a Droga , Comportamento Alimentar/fisiologia , Feminino , Privação de Alimentos/fisiologia , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/metabolismo , Imuno-Histoquímica , Injeções Intraventriculares , Neuropeptídeo Y/administração & dosagem , Distribuição Tecidual
11.
Brain Res ; 1184: 186-92, 2007 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-17980862

RESUMO

The correlation between male sexual behavior and catecholamine levels in the medial preoptic area (MPOA) and arcuate nucleus (ARN) was studied in middle-aged rats. Male rats (18-19 months) were assigned to three groups: (1) Group MIE, consisting of rats showing mounts, intromissions, and ejaculations; (2) Group MI, consisting of rats showing mounts and intromissions, but no ejaculation; and (3) Group NC, consisting of non-copulators showing no sexual behavior. Young adult rats (4-5 months) displaying complete copulatory behavior were used as the control group. Dopamine (DA) and norepinephrine (NE) tissue levels in the MPOA and ARN were measured by high pressure liquid chromatography with electrochemical detection. There were no differences between MIE rats and young controls in DA or NE tissue levels in these two brain areas. Furthermore, no differences were found between the MI and NC groups in DA or NE tissue levels in either the MPOA or ARN. DA tissue levels in the MPOA and ARN in the MI and NC groups were significantly lower than those in the MIE group. NE tissue levels in the MPOA of the NC group were significantly lower than those in the MIE group, but no differences in NE tissue levels in the ARN were seen between the four groups. These results suggest that, in male rats, complete male sexual performance is related to tissue levels of DA, but not of NE, in the MPOA and/or ARN. Furthermore, ejaculatory behavior might be associated with critical DA tissue levels in the MPOA and/or ARN in middle-aged rats.


Assuntos
Núcleo Arqueado do Hipotálamo/fisiologia , Catecolaminas/metabolismo , Área Pré-Óptica/fisiologia , Comportamento Sexual Animal/fisiologia , Fatores Etários , Análise de Variância , Animais , Comportamento Animal , Masculino , Ratos , Ratos Long-Evans , Estatísticas não Paramétricas
12.
Pharmacol Biochem Behav ; 83(2): 265-70, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16529800

RESUMO

The correlation between monoamine levels in the nucleus accumbens (NAcc) and male sexual behavior was studied in middle-aged rats. Male rats (18-19months) were assigned to three groups: (1) Group MIE consisted of rats showing mounts, intromissions, and ejaculations; (2) Group MI was composed of rats showing mounts and intromissions, but no ejaculation; and (3) Group NC were non-copulators showing no sexual behavior. Young adult rats (4-5months), displaying complete copulatory behavior, were used as the control group. Levels of dopamine (DA), serotonin, and norepinephrine and their metabolites in the NAcc were measured by high-pressure liquid chromatography with electrochemical detection. No difference was seen in DA levels between MIE rats and young controls, whereas DA levels in NC rats were significantly lower than those in both MIE and MI rats. Serotonin levels in NC rats were significantly higher than those in MIE and MI rats. Conversely, norepinephrine levels in NC rats were lower than those in MIE rats. These results suggest that monoamine levels in the NAcc correlate with sexual performance in male rats and that changes in NAcc monoamine levels might affect male sexual behavior in middle-aged rats.


Assuntos
Monoaminas Biogênicas/metabolismo , Núcleo Accumbens/metabolismo , Comportamento Sexual Animal , Animais , Cromatografia Líquida de Alta Pressão , Dopamina/metabolismo , Eletroquímica , Masculino , Ratos , Ratos Long-Evans , Serotonina/metabolismo
13.
Biol Reprod ; 71(3): 845-52, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15163616

RESUMO

The present study examined the gonadotropin surge-inducing actions of estradiol (E(2)), both alone and with progesterone (P(4)), in middle-aged, early persistent-estrous (PE) female rats that had become PE within 35 days. In addition, we also assessed the effect of P(4) on the mating-induced gonadotropin surges in these acyclic animals. Early PE rats were ovariectomized and received E(2) implants (Day 0). On Day 4, an s.c. injection of P(4) (0.5 mg/ 100 g body weight) at 1200 h markedly increased plasma P(4) and elicited both LH and FSH surges, whereas vehicle-treated controls displayed no rise in P(4) or gonadotropins. This observation confirms that at middle age, female rats no longer respond to the positive-feedback stimulation of E(2) on gonadotropin surges whenever the estrous cyclicity ceases. As PE continued, such a surge-inducing action of E(2) plus P(4) became diminished after 75 days of PE and disappeared thereafter. When caged with males, vehicle-treated early PE rats display a mating-induced increase in P(4) from the adrenal along with small gonadotropin surges. The amplitude of these mating-induced gonadotropin surges was enhanced by supplementation with exogenous P(4) in early PE rats. Our findings indicate that during the early phase of PE, the surge-inducing action of E(2) and P(4) remains intact but deteriorates as PE continues. Thus, a deficiency in P(4) secretion during aging may contribute to the diminished gonadotropin surge response in the hypothalamic-pituitary axis and the subsequent cessation of estrous cyclicity.


Assuntos
Envelhecimento/fisiologia , Hormônio Foliculoestimulante/sangue , Hormônio Luteinizante/sangue , Sistemas Neurossecretores/efeitos dos fármacos , Progesterona/farmacologia , Animais , Implantes de Medicamento , Estradiol/sangue , Estradiol/farmacologia , Feminino , Hormônio Foliculoestimulante/metabolismo , Hormônio Luteinizante/metabolismo , Masculino , Sistemas Neurossecretores/fisiologia , Ovariectomia , Postura , Progesterona/sangue , Ratos , Ratos Long-Evans , Comportamento Sexual Animal/efeitos dos fármacos
14.
Endocrinology ; 145(6): 2730-8, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15033909

RESUMO

17beta-Estradiol (E2)-stimulated estrogen receptor (ERalpha) transcription is accompanied by protein degradation via the 26S-proteasome pathway. Inhibition of proteasome activity stabilizes ERalpha protein and abolishes E2-activated transcription, suggesting functional linkages between transcription and degradation. It is not known whether ligand-independent ERalpha activation is coupled to proteolysis. In pituitary cells, forskolin (FSK) stimulates ERalpha transcription through the protein kinase A (PKA) pathway. This study examined interactions between E2-dependent and PKA-stimulated pathways in GH(3) cells by measuring transcription of a transfected reporter gene and endogenous ERalpha levels. E2 stimulated estrogen response element-mediated transcription 2- to 3-fold and decreased ERalpha protein levels to 40%. In contrast, FSK stimulated ERalpha transcription without decreasing ERalpha protein. Treatment with FSK plus E2 resulted in synergistic ERalpha transactivation, and FSK specifically prevented E2-induced ERalpha degradation. PKA is required for protection and was prevented by H89 (a PKA inhibitor), but not PD98059 (a MAPK kinase inhibitor). Propyl-pyrazole-triol and R,R-diethyl-tetrahydrochrysene, selective ERalpha agonists, reduced ERalpha protein by 50% while stimulating ERalpha transcriptional activity 4- to 8-fold. The antagonist ICI 182,780 similarly decreased ERalpha levels, but prevented ER activation. FSK prevented all ligand-induced ERalpha degradation. Lactacystin, a proteasome inhibitor, abolished E2-stimulated, but not FSK-stimulated, ERalpha transcription. Thus, stimulation of ERalpha transcription by the PKA-dependent pathway is dissociated from receptor degradation and proteasome activity. These data suggest a mechanism of ERalpha transcriptional activation by PKA that is distinct from E2 activation and that may contribute to the synergistic transcriptional activation of ERalpha by ligand-dependent and PKA-dependent pathways.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Cisteína Endopeptidases/fisiologia , Estradiol/análogos & derivados , Complexos Multienzimáticos/fisiologia , Receptores de Estrogênio/metabolismo , Animais , Linhagem Celular , Colforsina/farmacologia , Sinergismo Farmacológico , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio , Fulvestranto , Ligantes , Complexo de Endopeptidases do Proteassoma , Ratos , Receptores de Estrogênio/efeitos dos fármacos , Receptores de Estrogênio/genética , Transcrição Gênica/efeitos dos fármacos
15.
Biol Reprod ; 66(4): 1104-10, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11906931

RESUMO

Chronic exposure of young ovariectomized rats to elevated circulating estradiol causes loss of steroid-induced LH surges. Such LH surges are associated with cFos-induced activation of GnRH neurons; therefore, we hypothesized that chronic estradiol treatment abolishes LH surges by decreasing activation of GnRH neurons. Regularly cycling rats were ovariectomized and immediately received an estradiol implant or remained untreated. Three days or 2 or 4 wk later, the estradiol-treated rats received vehicle or progesterone at 1200 h, and 7 hourly blood samples were collected for RIA of LH. Thereafter, all rats were perfused, and the brains were examined for immunocytochemical localization of cFos and GnRH. The GnRH neurons from untreated ovariectomized rats rarely expressed cFos. As reported, LH surges induced by 3 days of estradiol treatment were associated with a 30% increase in cFos-containing GnRH neurons, and progesterone enhanced both the amplitude of LH surges and the proportion of cFos-immunopositive GnRH neurons. As hypothesized, the abolition of LH surges caused by 2 or more weeks of estradiol was paralleled by a reduction in the percentage of cFos-containing GnRH neurons, and this effect was delayed by progesterone. These results suggest that chronic estradiol abolishes steroid-induced LH surges in part by inactivating GnRH neurons.


Assuntos
Estradiol/administração & dosagem , Hormônio Liberador de Gonadotropina/análise , Hormônio Luteinizante/metabolismo , Neurônios/efeitos dos fármacos , Envelhecimento , Animais , Peso Corporal , Encéfalo/citologia , Química Encefálica , Implantes de Medicamento , Estradiol/sangue , Feminino , Hormônio Liberador de Gonadotropina/fisiologia , Hormônio Luteinizante/sangue , Neurônios/química , Neurônios/fisiologia , Ovariectomia , Progesterona/administração & dosagem , Proteínas Proto-Oncogênicas c-fos/análise , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA