Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Artigo em Inglês | MEDLINE | ID: mdl-29515521

RESUMO

Gonadotropin-releasing hormone (GnRH) neurons originate outside the central nervous system (CNS) in the nasal placode where their migration to the basal forebrain is dependent on the integration of multiple signaling cues during development. The proper migration and establishment of the GnRH neuronal population within the CNS are critical for normal pubertal onset and reproductive function. The endopeptidase EP24.15 is expressed along the migratory path of GnRH neurons and cleaves the full-length GnRH to generate the metabolite GnRH-(1-5). Using the GN11 cell model, which is considered a pre-migratory GnRH neuronal cell line, we demonstrated that GnRH-(1-5) inhibits cellular migration in a wound closure assay by binding the orphan G protein-coupled receptor 173 (GPR173). In our current experiments, we sought to utilize an in vitro migration assay that better reflects the external environment that migrating GnRH neurons are exposed to during development. Therefore, we used a transwell assay where the inserts were coated with or without a matrigel, a gelatinous mixture containing extracellular matrix (ECM) proteins, to mimic the extracellular environment. Interestingly, GnRH-(1-5) inhibited the ability of GN11 cells to migrate only through ECM mimetic and was dependent on GPR173. Furthermore, we found that GN11 cells secrete TGF-ß1, 2, and 3 but only TGF-ß1 release and signaling were inhibited by GnRH-(1-5). To identify potential mechanisms involved in the proteolytic activation of TGF-ß, we measured a panel of genes implicated in ECM remodeling. We found that GnRH-(1-5) consistently increased tissue inhibitors of metalloproteinase 1 expression, which is an inhibitor of proteinase activity, leading to a decrease in bioactive TGF-ß and subsequent signaling. These results suggest that GnRH-(1-5) activating GPR173 may modulate the response of migrating GnRH neurons to external cues present in the ECM environment via an autocrine-dependent mechanism involving TGF-ß.

3.
Behav Neurosci ; 130(6): 624-34, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27786501

RESUMO

Sexual receptivity in female rodents induced by the sequential injection of estrogen and progesterone is followed by a period in which females do not respond behaviorally to a second administration of progesterone (P); this is known as sequential inhibition. It has been proposed that the induction of sequential inhibition by progesterone in rats depends on down regulation of the progesterone receptor (PR) in brain areas involved in the expression of female sexual receptivity. P is rapidly metabolized to a variety of 5α- or 5ß-ring A-reduced progestins (RPrg). These RPrg have little or no affinity for the PR. They stimulate sexual receptivity (lordosis) more potently than P itself in estrogen-primed rats and do not induce sequential inhibition. The purpose of the current study was to test the role of the PR in the facilitation of lordosis and sequential inhibition induced by P and the following RPrg: 5α-pregnandione (5α-DHP), 5α,3ß-pregnanolone (5α,3ß-Pgl), 5ß-pregnanedione (5ß-DHP), and 5ß,3α-pregnanolone (5ß,3α-Pgl) in ovariectomized (ovx) female mice primed with estradiol benzoate. The RPrg were tested in C57BL/6 mice and in a strain lacking the progesterone receptor expression (PRKO). Our results show that both facilitation and sequential inhibition of lordosis induced by progesterone require the presence of the progesterone receptor. Interestingly, some RPrg facilitate lordosis but do not induce sequential inhibition in female mice. Sexual receptivity induced by RPrg does not require the progesterone receptor. Thus, RPrg induce sexual receptivity, but they probably exert their effects through a different cellular mechanism that does not involve the progesterone receptor. (PsycINFO Database Record


Assuntos
Progesterona/metabolismo , Progestinas/metabolismo , Receptores de Progesterona , Animais , Estradiol/análogos & derivados , Estrogênios , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Ovariectomia , Pregnanolona , Comportamento Sexual Animal/fisiologia
5.
Artigo em Inglês | MEDLINE | ID: mdl-26528239

RESUMO

Oxytocin (OT) is a neuropeptide synthesized primarily by neurons of the paraventricular and supraoptic nuclei of the hypothalamus. These neurons have axons that project into the posterior pituitary and release OT into the bloodstream to promote labor and lactation; however, OT neurons also project to other brain areas where it plays a role in numerous brain functions. OT binds to the widely expressed OT receptor (OTR), and, in doing so, it regulates homeostatic processes, social recognition, and fear conditioning. In addition to these functions, OT decreases neuroendocrine stress signaling and anxiety-related and depression-like behaviors. Steroid hormones differentially modulate stress responses and alter OTR expression. In particular, estrogen receptor ß activation has been found to both reduce anxiety-related behaviors and increase OT peptide transcription, suggesting a role for OT in this estrogen receptor ß-mediated anxiolytic effect. Further research is needed to identify modulators of OT signaling and the pathways utilized and to elucidate molecular mechanisms controlling OT expression to allow better therapeutic manipulations of this system in patient populations.

6.
eNeuro ; 2(4)2015.
Artigo em Inglês | MEDLINE | ID: mdl-26465008

RESUMO

Estrogens act in brain to profoundly influence neurogenesis, sexual differentiation, neuroprotection, cognition, energy homeostasis, and female reproductive behavior and physiology through a variety of mechanisms, including the induction of progestin receptors (PRs). PRs are expressed as two isoforms, PR-A and PR-B, that have distinct functions in physiology and behavior. Because these PR isoforms cannot be distinguished using cellular resolution techniques, the present study used isoform-specific null mutant mice that lack PR-A or PR-B for the first time to investigate whether 17ß-estradiol benzoate (EB) regulates the differential expression of the PR isoforms in the ventromedial nucleus of the hypothalamus (VMN), arcuate nucleus, and medial preoptic area, brain regions that are rich in EB-induced PRs. Interestingly, EB induced more PR-A than PR-B in all three brain regions, suggesting that PR-A is the predominant isoform in these regions. Given that steroid receptor coactivator (SRC)-1 and SRC-2 are important in estrogen receptor (ER)-dependent transcription in brain, including PR induction, we tested whether the expression of these coactivators was correlated with PR isoform expression. The majority of EB-induced PR cells expressed both SRC-1 and SRC-2 in the three brain regions of all genotypes. Interestingly, the intensity of PR-A immunoreactivity correlated with SRC-2 expression in the VMN, providing a potential mechanism for selective ER-mediated transactivation of PR-A over PR-B in a brain region-specific manner. In summary, these novel findings indicate that estrogens differentially regulate PR-A and PR-B expression in the female hypothalamus, and provide a mechanism by which steroid action in brain can selectively modulate behavior and physiology.

7.
Mol Cell Endocrinol ; 415: 114-25, 2015 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-26277400

RESUMO

In the extracellular space, the gonadotropin-releasing hormone (GnRH) is metabolized by the zinc metalloendopeptidase EC3.4.24.15 (EP24.15) to form the pentapeptide, GnRH-(1-5). GnRH-(1-5) diverges in function and mechanism of action from GnRH in the brain and periphery. GnRH-(1-5) acts on the orphan G protein-coupled receptor 101 (GPR101) to sequentially stimulate epidermal growth factor (EGF) release, phosphorylate the EGF receptor (EGFR), and facilitate cellular migration. These GnRH-(1-5) actions are dependent on matrix metallopeptidase (MMP) activity. Here, we demonstrated that these GnRH-(1-5) effects are dependent on increased MMP-9 enzymatic activity in the Ishikawa and ECC-1 cell lines. Furthermore, the effects of GnRH-(1-5) mediated by GPR101 and the subsequent increase in MMP-9 enzymatic activity lead to an increase in cellular invasion. These results suggest that GnRH-(1-5) and GPR101 regulation of MMP-9 may have physiological relevance in the metastatic potential of endometrial cancer cells.


Assuntos
Neoplasias do Endométrio/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Fragmentos de Peptídeos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Metástase Neoplásica , Fosforilação , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais
8.
Endocrinology ; 156(6): 2150-61, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25849728

RESUMO

In addition to androgenic properties mediated via androgen receptors, dihydrotestosterone (DHT) also regulates estrogenic functions via an alternate pathway. These estrogenic functions of DHT are mediated by its metabolite 5α-androstane-3ß, 17ß-diol (3ß-diol) binding to estrogen receptor ß (ERß). CYP7B1 enzyme converts 3ß-diol to inactive 6α- or 7α-triols and plays an important role as a regulator of estrogenic functions mediated by 3ß-diol. Using a mutant mouse carrying a null mutation for the CYP7B1 gene (CYP7B1KO), we examined the contribution of CYP7B1 on physiology and behavior. Male, gonadectomized (GDX) CYP7B1KO and their wild type (WT) littermates were assessed for their behavioral phenotype, anxiety-related behavioral measures, and hypothalamic pituitary adrenal axis reactivity. No significant effects of genotype were evident in anxiety-like behaviors in open field (OFA), light-dark (L/D) exploration, and elevated plus maze (EPM). T significantly reduced open arm time on the EPM while not affecting L/D exploratory and OFA behaviors in CYP7B1KO and WT littermates. T also attenuated the corticosterone response to EPM in both genotypes. In GDX animals, T was able to reinstate male-specific reproductive behaviors (latencies and number of mounts, intromission, and ejaculations) in the WT but not in the CYP7B1KO mice. The male reproductive behavior defect in CYP7B1KO seems to be due to their inability to distinguish olfactory cues from a behavioral estrus female. CYP7B1KO mice also showed a reduction in androgen receptor mRNA expression in the olfactory bulb. Our findings suggest a novel role for the CYP7B1 enzyme in the regulation of male reproductive behaviors.


Assuntos
Comportamento Sexual Animal/fisiologia , Esteroide Hidroxilases/fisiologia , Animais , Ansiedade/genética , Ansiedade/metabolismo , Ansiedade/fisiopatologia , Calbindinas/genética , Calbindinas/metabolismo , Calbindinas/fisiologia , Família 7 do Citocromo P450 , Feminino , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Esteroide Hidroxilases/genética , Esteroide Hidroxilases/metabolismo
9.
J Clin Invest ; 124(10): 4351-62, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25157819

RESUMO

Binge eating afflicts approximately 5% of US adults, though effective treatments are limited. Here, we showed that estrogen replacement substantially suppresses binge-like eating behavior in ovariectomized female mice. Estrogen-dependent inhibition of binge-like eating was blocked in female mice specifically lacking estrogen receptor-α (ERα) in serotonin (5-HT) neurons in the dorsal raphe nuclei (DRN). Administration of a recently developed glucagon-like peptide-1-estrogen (GLP-1-estrogen) conjugate designed to deliver estrogen to GLP1 receptor-enhanced regions effectively targeted bioactive estrogens to the DRN and substantially suppressed binge-like eating in ovariectomized female mice. Administration of GLP-1 alone reduced binge-like eating, but not to the same extent as the GLP-1-estrogen conjugate. Administration of ERα-selective agonist propylpyrazole triol (PPT) to murine DRN 5-HT neurons activated these neurons in an ERα-dependent manner. PPT also inhibited a small conductance Ca2+-activated K+ (SK) current; blockade of the SK current prevented PPT-induced activation of DRN 5-HT neurons. Furthermore, local inhibition of the SK current in the DRN markedly suppressed binge-like eating in female mice. Together, our data indicate that estrogens act upon ERα to inhibit the SK current in DRN 5-HT neurons, thereby activating these neurons to suppress binge-like eating behavior and suggest ERα and/or SK current in DRN 5-HT neurons as potential targets for anti-binge therapies.


Assuntos
Estrogênios/metabolismo , Comportamento Alimentar/efeitos dos fármacos , Neurônios/metabolismo , Serotonina/metabolismo , Animais , Transtorno da Compulsão Alimentar , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/genética , Feminino , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Núcleos da Rafe/metabolismo
11.
Mol Endocrinol ; 28(1): 80-98, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24264576

RESUMO

The decapeptide GnRH is known for its central role in the regulation of the hypothalamo-pituitary-gonadal axis. In addition, it is also known to have local effects within peripheral tissues. The zinc metalloendopeptidase, EC 3.4.24.15 (EP24.15), can cleave GnRH at the Tyr(5)-Gly(6) bond to form the pentapeptide, GnRH-(1-5). The central and peripheral effect of GnRH-(1-5) is different from its parent peptide, GnRH. In the current study, we examined the effect of GnRH-(1-5) on epidermal growth factor receptor (EGFR) phosphorylation and cellular migration. Using the Ishikawa cell line as a model of endometrial cancer, we demonstrate that GnRH-(1-5) stimulates epidermal growth factor release, increases the phosphorylation of EGFR (P < .05) at three tyrosine sites (992, 1045, 1068), and promotes cellular migration. In addition, we also demonstrate that these actions of GnRH-(1-5) are mediated by the orphan G protein-coupled receptor 101 (GPR101). Down-regulation of GPR101 expression blocked the GnRH-(1-5)-mediated release of epidermal growth factor and the subsequent phosphorylation of EGFR and cellular migration. These results suggest that GPR101 is a critical requirement for GnRH-(1-5) transactivation of EGFR in Ishikawa cells.


Assuntos
Receptores ErbB/genética , Hormônio Liberador de Gonadotropina/fisiologia , Proteínas Oncogênicas/metabolismo , Fragmentos de Peptídeos/fisiologia , Receptores de Superfície Celular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Ativação Transcricional , Sinalização do Cálcio , Linhagem Celular Tumoral , Movimento Celular , Neoplasias do Endométrio , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Feminino , Expressão Gênica , Humanos , Inibidores de Metaloproteinases de Matriz/farmacologia , Oligopeptídeos/fisiologia , Proteínas Oncogênicas/genética , Fenilalanina/análogos & derivados , Fenilalanina/farmacologia , Fosforilação , Processamento de Proteína Pós-Traducional , Ácido Pirrolidonocarboxílico/análogos & derivados , Quinazolinas/farmacologia , Receptores de Superfície Celular/genética , Receptores Acoplados a Proteínas G/genética , Receptores LHRH , Tiofenos/farmacologia , Tirfostinas/farmacologia
12.
Endocrinology ; 154(12): 4726-36, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24140715

RESUMO

We have previously demonstrated that the cleavage product of the full-length GnRH, GnRH-(1-5), is biologically active, binds G protein-coupled receptor 173 (GPR173), and inhibits the migration of cells in the immortalized GnRH-secreting GN11 cell. In this study, we attempted to characterize the GnRH-(1-5) intracellular signaling mechanism. To determine whether the signaling pathway mediating GnRH-(1-5) regulation of migration involves a G protein-dependent mechanism, cells were treated with a generic G protein antagonist in the presence and absence of GnRH-(1-5), and a wound-healing assay was conducted to measure migration. G Protein antagonist 2 treatment abolished the GnRH-(1-5) inhibition of migration, indicating that the mechanism of GnRH-(1-5) is G protein coupled. To identify the potential Gα-subunit recruited by GnRH-(1-5) binding GPR173, we measured the second messengers cAMP and inositol triphosphate levels. GnRH-(1-5) treatment did not alter cAMP levels relative to cells treated with vehicle or forskolin, suggesting that GnRH-(1-5) does not couple to the Gαs or Gαi subunits. Similarly, inositol triphosphate levels remained unchanged with GnRH-(1-5) treatment, indicating a mechanism not mediated by the Gαq/11 subunit. Therefore, we also examined whether GnRH-(1-5) activating GPR173 deviated from the canonical G protein-coupled receptor signaling pathway by coupling to ß-arrestin 1/2 to regulate migration. Our coimmunoprecipitation studies indicate that GnRH-(1-5) induces the rapid interaction between GPR173 and ß-arrestin 2 in GN11 cells. Furthermore, we demonstrate that this association recruits phosphatase and tensin homolog to mediate the downstream action of GnRH-(1-5). These findings suggest that the GnRH-(1-5) mechanism deviates from the canonical G protein-coupled receptor pathway to regulate cell migration in immortalized GnRH neurons.


Assuntos
Arrestinas/metabolismo , Hormônio Liberador de Gonadotropina/farmacologia , Neurônios/efeitos dos fármacos , Animais , Arrestinas/genética , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , AMP Cíclico , Hormônio Liberador de Gonadotropina/química , Hormônio Liberador de Gonadotropina/metabolismo , Inositol 1,4,5-Trifosfato/metabolismo , Camundongos , Neurônios/citologia , Neurônios/fisiologia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , beta-Arrestina 1 , beta-Arrestina 2 , beta-Arrestinas
13.
Artigo em Inglês | MEDLINE | ID: mdl-23847594

RESUMO

The gonadotropin-releasing hormone (GnRH) was originally isolated from the mammalian hypothalamus for its role as the primary regulator of reproductive function. Since its discovery, GnRH has also been shown to be located in non-hypothalamic tissues and is known to have diverse functions. Although the regulation of GnRH synthesis and release has been extensively studied, there is additional evidence to suggest that the processing of GnRH to the metabolite GnRH-(1-5) represents another layer of regulation. The focus of this review will be on the current evidence for the action of the pentapeptide metabolite GnRH-(1-5) in regulating cellular migration. We discuss the potential role of GnRH-(1-5) in regulating GnRH neuronal migration during development. Furthermore, we demonstrate these actions are mediated by the activation of a G protein-coupled receptor. Our findings suggest that GnRH-(1-5) may play a developmental function in addition to regulating developing cells.

14.
Endocrinology ; 154(2): 783-95, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23321696

RESUMO

The decapeptide GnRH is an important regulator of reproductive behavior and function. In the extracellular matrix, GnRH is metabolized by the endopeptidase EC3.4.24.15 (EP24.15) to generate the pentapeptide GnRH-(1-5). In addition to its expression in the adult hypothalamus, EP24.15 is expressed along the migratory path of GnRH-expressing neurons during development. Although we have previously demonstrated a role for EP24.15 in the generation of the biologically active pentapeptide GnRH-(1-5) in regulating GnRH expression and mediating sexual behavior during adulthood in rodents, the modulatory role of GnRH-(1-5) in the migration of GnRH neurons during development remains unknown. To address this information gap, we examined the effect of GnRH-(1-5) on the cellular migration of a premigratory GnRH-secreting neuronal cell line, the GN11 cell, using a wound-healing assay. Dose- and time-response studies demonstrated that GnRH-(1-5) significantly delayed wound closure. We then sought to identify the mechanism by which GnRH-(1-5) inhibits migration. Because the cognate GnRH receptor is a G protein-coupled receptor, we examined whether GnRH-(1-5) regulates migration by also activating a G protein-coupled receptor. Using a high-throughput ß-arrestin recruitment assay, we identified an orphan G protein-coupled receptor (GPR173) that was specifically activated by GnRH-(1-5). Interestingly, small interfering RNA to GPR173 reversed the GnRH-(1-5)-mediated inhibition on migration of GN11 neurons. Furthermore, we also demonstrate that the GnRH-(1-5)-activated GPR173-dependent signal transduction pathway involves the activation of the signal transducer and activator of transcription 3 in GnRH migration. These findings indicate a potential regulatory role for GnRH-(1-5) in GnRH neuronal migration during development.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Fragmentos de Peptídeos/farmacologia , Receptores Acoplados a Proteínas G/fisiologia , Animais , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/farmacologia , Masculino , Metaloendopeptidases/metabolismo , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/fisiologia , Cicatrização/efeitos dos fármacos
15.
Artigo em Inglês | MEDLINE | ID: mdl-22649404

RESUMO

Steroid hormone, progesterone, modulates neuroendocrine functions in the central nervous system resulting in alterations in physiology and behavior. These neuronal effects are mediated primarily by intracellular progestin receptors (PRs) in the steroid-sensitive neurons, resulting in transcription-dependent genomic actions (classical mechanism). In addition to progesterone, intracellular PRs can also be activated in a "ligand-independent" manner by neurotransmitters, peptide growth factors, cyclic nucleotides, and neurosteroids. Recent studies indicate that rapid, non-classical progesterone actions involving cytoplasmic kinase signaling and/or extranuclear PRs can result in both transcription-independent and transcription-dependent actions. Cross-talk between extranuclear and classical intracellular signaling pathways promotes progesterone-dependent behavior in mammals. This review focuses on the mechanisms by which progesterone-initiated signaling mechanisms converge with PRs in the brain to modulate reproductive behavior in female rodents.

16.
Neuroendocrinology ; 96(2): 111-8, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22538291

RESUMO

It is now well established that estrogens can influence a panoply of physiological and behavioral functions. In many instances, the effects of estrogens are mediated by the 'classical' actions of two different estrogen receptors (ERs), ERα or ERß. ERα and ERß appear to have opposing actions in the control of stress responses and modulate different neurotransmitter or neuropeptide systems. Studies elucidating the molecular mechanisms for such regulatory processes are currently in progress. Furthermore, the use of ERα and ERß knockout mouse lines has allowed the exploration of the importance of these receptors in behavioral responses such as anxiety-like and depressive-like behaviors. This review examines some of the recent advances in our knowledge of hormonal control of neuroendocrine and behavioral responses to stress and underscore the importance of these receptors as future therapeutic targets for control of stress-related signaling pathways.


Assuntos
Estrogênios/metabolismo , Sistemas Neurossecretores/fisiologia , Receptores de Estrogênio/metabolismo , Estresse Psicológico/patologia , Animais , Estrogênios/farmacologia , Humanos , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia
17.
Neuroendocrinology ; 96(2): 152-61, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22538437

RESUMO

The steroid hormone, progesterone (P), modulates neuroendocrine functions in the central nervous system resulting in integration of reproduction and reproductive behaviors in female mammals. Although it is widely recognized that P's effects on female sex behavior are mediated by the classical neural progestin receptors (PRs) functioning as 'ligand-dependent' transcription factors to regulate genes and genomic networks, additional mechanisms of PR activation also contribute to the behavioral response. Cellular and molecular evidence indicates that PRs can be activated in a ligand-independent manner by neurotransmitters, growth factors, cyclic nucleotides, progestin metabolites and mating stimuli. The rapid responses of P may be mediated by a variety of PR types, including membrane-associated PRs or extranuclear PRs. Furthermore, these rapid nonclassical P actions involving cytoplasmic kinase signaling and/or extranuclear PRs also converge with classical PR-mediated transcription-dependent pathways to regulate reproductive behaviors. In this review, we summarize some of the history of the study of the role of PRs in reproductive behaviors and update the status of PR-mediated mechanisms involved in the facilitation of female sex behavior. We present an integrative model of PR activation via crosstalk and convergence of multiple signaling pathways.


Assuntos
Encéfalo/metabolismo , Receptores de Progesterona/metabolismo , Caracteres Sexuais , Comportamento Sexual/fisiologia , Animais , Feminino , Humanos
18.
Endocrinology ; 153(2): 837-46, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22186418

RESUMO

The dichotomous anxiogenic and anxiolytic properties of estrogens have been reported to be mediated by two distinct neural estrogen receptors (ER), ERα and ERß, respectively. Using a combination of pharmacological and genetic approaches, we confirmed that the anxiolytic actions of estradiol are mediated by ERß and extended and these observations to demonstrate the neuroanatomical targets involved in ERß activation in these behavioral responses. We examined the effects of the biologically active S-enantiomer of diarylpropionitrile (S-DPN) on anxiety-related behavioral measures, the corresponding stress hormonal response to hypothalamo-pituitary-adrenal axis reactivity, and potential sites of neuronal activation in mutant female mice carrying a null mutation for ERß gene (ßERKO). S-DPN administration significantly reduced anxiety-like behaviors in the open field, light-dark exploration, and the elevated plus maze (EPM) in ovariectomized wild-type (WT) mice, but not in their ßERKO littermates. Stress-induced corticosterone (CORT) and ACTH were also attenuated by S-DPN in the WT mice but not in the ßERKO mice. Using c-fos induction after elevated plus maze, as a marker of stress-induced neuronal activation, we identified the anterodorsal medial amygdala and bed nucleus of the stria terminalis as the neuronal targets of S-DPN action. Both areas showed elevated c-fos mRNA expression with S-DPN treatment in the WT but not ßERKO females. These studies provide compelling evidence for anxiolytic effects mediated by ERß, and its neuroanatomical targets, that send or receive projections to/from the paraventricular nucleus, providing potential indirect mode of action for the control of hypothalamo-pituitary-adrenal axis function and behaviors.


Assuntos
Ansiolíticos/farmacologia , Comportamento Animal/fisiologia , Receptor beta de Estrogênio/agonistas , beta-Ciclodextrinas/farmacologia , 2-Hidroxipropil-beta-Ciclodextrina , Animais , Ansiedade/metabolismo , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Feminino , Regulação da Expressão Gênica/fisiologia , Camundongos , Camundongos Knockout , Mutação , Nitrilas/farmacologia , Ovariectomia , Propionatos/farmacologia , Estresse Fisiológico
19.
Front Neuroendocrinol ; 31(2): 157-71, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20116396

RESUMO

The steroid hormone, progesterone (P), modulates neuroendocrine functions in the central nervous system resulting in alterations in physiology and reproductive behavior in female mammals. A wide body of evidence indicates that these neural effects of P are predominantly mediated via their intracellular progestin receptors (PRs) functioning as "ligand-dependent" transcription factors in the steroid-sensitive neurons regulating genes and genomic networks. In addition to P, intracellular PRs can be activated by neurotransmitters, growth factors and cyclic nucleotides in a ligand-independent manner via crosstalk and convergence of pathways. Furthermore, recent studies indicate that rapid signaling events associated with membrane PRs and/or extra-nuclear, cytoplasmic PRs converge with classical PR activated pathways in neuroendocrine regulation of female reproductive behavior. The molecular mechanisms, by which multiple signaling pathways converge on PRs to modulate PR-dependent female reproductive behavior, are discussed in this review.


Assuntos
Neurotransmissores/metabolismo , Receptores de Progesterona/fisiologia , Comportamento Sexual Animal/fisiologia , Animais , Dopamina/fisiologia , Estrogênios/farmacologia , Feminino , Ligantes , Fosforilação , Receptores de Progesterona/efeitos dos fármacos , Receptores de Progesterona/metabolismo , Transdução de Sinais/fisiologia
20.
Endocrinology ; 149(11): 5518-26, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18617607

RESUMO

In addition to the activation of classical progestin receptor-dependent genomic pathway, progesterone (P) can activate nonclassical, membrane-initiated signaling pathways in the brain. We recently demonstrated rapid P activation of second-messenger kinases, protein kinase A, and protein kinase C in the ventromedial nucleus (VMN) and preoptic area (POA) of rat brain. To determine whether P can activate yet another Ca+2 dependent kinase, we examined the rapid P modulation of calcium and calmodulin-dependent protein kinase II (CaMKII) in the VMN and POA in female rats. A rapid P-initiated activation of CaMKII basal activity was observed in the VMN but not the POA at 30 min. Estradiol benzoate (EB) priming enhanced this CaMKII basal activity in both the VMN and POA. CaMKII protein levels and phosphorylation of Thr-286 moiety on CaMKII, however, remained unchanged with EB and/or P treatments, suggesting that the changes in the CaMKII kinase activity are due to rapid P modulation of the kinase activity and not its synthesis or autoactivation. Furthermore, intracerebroventricular (icv) administration of a CaMKII-specific inhibitor, KN-93, 30 min prior to the P infusion, in EB-primed, ovariectomized female rats inhibited CaMKII activation but not protein kinase A and protein kinase C activities. Interestingly, icv administration of KN-93 30 min prior to P infusion (icv) resulted in a reduction but not total inhibition of P-facilitated lordosis response in EB-primed female rats. These observations suggest a redundancy or, alternately, a hierarchy in the P-regulated activation of kinase signaling cascades in female reproductive behavior.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/fisiologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/enzimologia , Progesterona/farmacologia , Animais , Benzilaminas/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/enzimologia , Ativação Enzimática/efeitos dos fármacos , Estradiol/análogos & derivados , Estradiol/farmacologia , Feminino , Hipotálamo/metabolismo , Ovariectomia , Fosforilação/efeitos dos fármacos , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/enzimologia , Área Pré-Óptica/metabolismo , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Sprague-Dawley , Comportamento Sexual Animal/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...