Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
J Neuroendocrinol ; 34(12): e13212, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36426781

RESUMO

The ventromedial prefrontal cortex (vmPFC) regulates fear acquisition, fear extinction, mood, and HPA axis function. Multiple brain regions exhibit time-of-day dependent variations in learning, long term potentiation (LTP), and dendritic morphology. Glucocorticoids have been implicated in the regulation of dendritic structure in the context of stress. Glucocorticoids are also known to regulate molecular clock entrainment via upregulation of Per1 transcription. In the present study, C57BL/6 N mice were sacrificed at three distinct times of day (ZT3, ZT12, and ZT16, lights off at ZT12) and Per1 mRNA expression was measured in the infralimbic and prelimbic vmPFC subregions using droplet digital (dd) PCR after recovering from adrenalectomy or sham surgery for 10 days. Sham mice showed Per1 rhythmicity in both infralimbic (IL) and prelimbic (PL) cortex, with peak expression occurring at ZT12. Adrenalectomized mice showed reductions in Per1 amplitude at ZT12 in both IL and PL, suggesting that the vmPFC molecular clock is entrained by diurnal glucocorticoid oscillations. Thy1-eGFP mice were used to visualize and quantify dendritic spine density on deep layer pyramidal dendrites at ZT 3, 12, and 16. Spine density in both PL and IL exhibited changes between the light (inactive) and dark (active) phases, with peak spine density observed at ZT16 and trough spine density observed at ZT3. These changes in spine density were restricted to changes in long thin and stubby type spines. To determine if changes in spine density is regulated by glucocorticoid oscillations, the 11ß-hydroxylase inhibitor metyrapone was administered 2 h prior to the onset of the active phase (ZT10) daily for 7 days. Metyrapone administration blocked both the diurnal peak of plasma corticosterone and peak spine densities in the IL and PL at ZT16. These results suggest that vmPFC molecular clock gene and dendritic spine diurnal rhythms depend on intact diurnal glucocorticoid oscillations.


Assuntos
Extinção Psicológica , Glucocorticoides , Animais , Camundongos , Ritmo Circadiano/fisiologia , Extinção Psicológica/fisiologia , Medo/fisiologia , Glucocorticoides/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Metirapona/farmacologia , Camundongos Endogâmicos C57BL , Sistema Hipófise-Suprarrenal/metabolismo , Córtex Pré-Frontal/metabolismo
2.
Artigo em Inglês | MEDLINE | ID: mdl-35667789

RESUMO

Sex differences in the neuroendocrine response to acute stress occur in both animals and humans. In rodents, stressors such as restraint and novelty induce a greater activation of the hypothalamic-pituitary-adrenal axis (HPA) in females compared to males. The nature of this difference arises from steroid actions during development (organizational effects) and adulthood (activational effects). Androgens decrease HPA stress responsivity to acute stress, while estradiol increases it. Androgenic down-regulation of HPA responsiveness is mediated by the binding of testosterone (T) and dihydrotestosterone (DHT) to the androgen receptor, as well as the binding of the DHT metabolite, 3ß-diol, to the ß form of the estrogen receptor (ERß). Estradiol binding to the α form of the estrogen receptor (ERα) increases HPA responsivity. Studies of human sex differences are relatively few and generally employ a psychosocial paradigm to measure stress-related HPA activation. Men consistently show greater HPA reactivity than women when being evaluated for achievement. Some studies have found greater reactivity in women when being evaluated for social performance. The pattern is inconsistent with rodent studies but may involve the differential nature of the stressors employed. Psychosocial stress is nonphysical and invokes a significant degree of top-down processing that is not easily comparable to the types of stressors employed in rodents. Gender identity may also be a factor based on recent work showing that it influences the neural processing of positive and negative emotional stimuli independent of genetic sex. Comparing different types of stressors and how they interact with gender identity and genetic sex will provide a better understanding of sex steroid influences on stress-related HPA reactivity.

3.
Androg Clin Res Ther ; 2(1): 261-274, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35024695

RESUMO

Androgens play a pivotal role during development. These gonadal hormones and their receptors exert organizational actions that shape brain morphology in regions controlling the stress regulatory systems in a male-specific manner. Specifically, androgens drive sex differences in the hypothalamic/pituitary/adrenal (HPA) axis and corresponding hypothalamic neuropeptides. While studies have examined the role of estradiol and its receptors in sex differences in the HPA axis and associated behaviors, the role of androgens remains far less studied. Androgens are generally thought to modulate the HPA axis through the activation of androgen receptors (ARs). They can also impact the HPA axis through reduction to estrogenic metabolites that can bind estrogen receptors in the brain and periphery. Such regulation of the HPA axis stress response by androgens can often result in sex-biased risk factors for stress-related disorders, such as anxiety and depression. This review focuses on the biosynthesis pathways and molecular actions of androgens and their nuclear receptors. The impact of androgens on hypothalamic neuropeptide systems (corticotropin-releasing hormone, arginine vasopressin, oxytocin, dopamine, and serotonin) that control the stress response and stress-related disorders is discussed. Finally, this review discusses potential therapeutics involving androgens (androgen replacement therapies, selective AR modulator therapies) and ongoing clinical trials.

4.
Biol Sex Differ ; 11(1): 44, 2020 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-32727567

RESUMO

Estradiol and testosterone are powerful steroid hormones that impact brain function in numerous ways. During development, these hormones can act to program the adult brain in a male or female direction. During adulthood, gonadal steroid hormones can activate or inhibit brain regions to modulate adult functions. Sex differences in behavioral and neuroendocrine (i.e., hypothalamic pituitary adrenal (HPA) axis) responses to stress arise as a result of these organizational and activational actions. The sex differences that are present in the HPA and behavioral responses to stress are particularly important considering their role in maintaining homeostasis. Furthermore, dysregulation of these systems can underlie the sex biases in risk for complex, stress-related diseases that are found in humans. Although many studies have explored the role of estrogen and estrogen receptors in mediating sex differences in stress-related behaviors and HPA function, much less consideration has been given to the role of androgens. While circulating androgens can act by binding and activating androgen receptors, they can also act by metabolism to estrogenic molecules to impact estrogen signaling in the brain and periphery. This review focuses on androgens as an important hormone for modulating the HPA axis and behaviors throughout life and for setting up sex differences in key stress regulatory systems that could impact risk for disease in adulthood. In particular, impacts of androgens on neuropeptide systems known to play key roles in HPA and behavioral responses to stress (corticotropin-releasing factor, vasopressin, and oxytocin) are discussed. A greater knowledge of androgen action in the brain is key to understanding the neurobiology of stress in both sexes.


Assuntos
Androgênios/metabolismo , Sistemas Neurossecretores/fisiologia , Estresse Fisiológico , Humanos , Sistema Hipotálamo-Hipofisário , Sistema Hipófise-Suprarrenal , Fatores Sexuais
5.
Endocrinology ; 161(1)2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31754709

RESUMO

To limit excessive glucocorticoid secretion following hypothalamic-pituitary-adrenal (HPA) axis stimulation, circulating glucocorticoids inhibit corticotropin-releasing hormone (CRH) expression in paraventricular nucleus (PVN) neurons. As HPA function differs between sexes and depends on circulating estradiol (E2) levels in females, we investigated sex/estrous stage-dependent glucocorticoid regulation of PVN Crh. Using NanoString nCounter technology, we first demonstrated that adrenalectomized (ADX'd) diestrous female (low E2), but not male or proestrous female (high E2), mice exhibited a robust decrease in PVN CRH mRNA following 2-day treatment with the glucocorticoid receptor (GR) agonist RU28362. Immunohistochemical analysis of PVN CRH neurons in Crh-IRES-Cre;Ai14 mice, where TdTomato fluorescence permanently tags CRH-expressing neurons, showed similarly abundant co-expression of GR-immunoreactivity in males, diestrous females, and proestrous females. However, we identified sex/estrous stage-related glucocorticoid regulation or expression of GR transcriptional coregulators. Out of 17 coregulator genes examined using nCounter multiplex analysis, mRNAs that were decreased by RU28362 in ADX'd mice in a sex/estrous stage-dependent fashion included: GR (males = diestrous females > proestrous females), signal transducer and activator of transcription 3 (STAT3) (males < diestrous = proestrous), and HDAC1 (males < diestrous > proestrous). Steroid receptor coactivator 3 (SRC-3), nuclear corepressor 1 (NCoR1), heterogeneous nuclear ribonucleoprotein U (hnrnpu), CREB binding protein (CBP) and CREB-regulated transcription coactivator 2 (CRTC2) mRNAs were lower in ADX'd diestrous and proestrous females versus males. Additionally, most PVN CRH neurons co-expressed methylated CpG binding protein 2 (MeCP2)-immunoreactivity in diestrous female and male Crh-IRES-Cre;Ai14 mice. Our findings collectively suggest that GR's sex-dependent regulation of PVN Crh may depend upon differences in the GR transcriptional machinery and an underlying influence of E2 levels in females.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Estradiol/sangue , Glucocorticoides/metabolismo , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Adrenalectomia , Androstanóis/farmacologia , Animais , Hormônio Liberador da Corticotropina/genética , Ciclo Estral , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Hormônio Liberador de Gonadotropina/agonistas , Hipotálamo/citologia , Masculino , Camundongos , Camundongos Endogâmicos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , RNA Mensageiro , Fatores Sexuais , Vagina/citologia
6.
Endocrinology ; 159(9): 3378-3388, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-30060079

RESUMO

Atrazine (ATR) is a commonly used pre-emergence and early postemergence herbicide. Rats gavaged with ATR and its chlorometabolites desethylatrazine (DEA) and deisopropylatrazine (DIA) respond with a rapid and dose-dependent rise in plasma corticosterone, whereas the major chlorometabolite, diaminochlorotriazine (DACT), has little or no effect on corticosterone levels. In this study, we investigated the possible sites of ATR activation of the hypothalamic-pituitary-adrenal (HPA) axis. ATR treatment had no effect on adrenal weights but altered adrenal morphology. Hypophysectomized rats or rats under dexamethasone suppression did not respond to ATR treatment, suggesting that ATR does not directly stimulate the adrenal gland to induce corticosterone synthesis. Immortalized mouse corticotrophs (AtT-20) and primary rat pituitary cultures were treated with ATR, DEA, DIA, or DACT. None of the compounds induced an increase in ACTH secretion or potentiated ACTH release in conjunction with CRH on ACTH release. In female rats gavaged with ATR, pretreatment with the CRH receptor antagonist astressin completely blocked the ATR-induced rise in corticosterone concentrations, implicating CRH release in ATR-induced HPA activation. Intracerebroventricular infusion of ATR, DEA, and DIA but not DACT at concentrations equivalent to peak plasma concentrations after gavage dosing resulted in an elevation of plasma corticosterone concentrations. However, ATR did not induce c-Fos immunoreactivity in the paraventricular nucleus of the hypothalamus. These results indicate that ATR activates the HPA axis centrally and requires CRH receptor activation, but it does not stimulate cellular pathways associated with CRH neuronal excitation.


Assuntos
Atrazina/farmacologia , Corticotrofos/efeitos dos fármacos , Herbicidas/farmacologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Hipófise/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Glândulas Suprarrenais/efeitos dos fármacos , Glândulas Suprarrenais/patologia , Hormônio Adrenocorticotrópico/efeitos dos fármacos , Hormônio Adrenocorticotrópico/metabolismo , Animais , Atrazina/análogos & derivados , Linhagem Celular , Corticosterona/metabolismo , Corticotrofos/metabolismo , Dexametasona/farmacologia , Feminino , Glucocorticoides/farmacologia , Sistema Hipotálamo-Hipofisário/metabolismo , Camundongos , Técnicas de Cultura de Órgãos , Tamanho do Órgão , Hipófise/metabolismo , Hipófise/cirurgia , Sistema Hipófise-Suprarrenal/metabolismo , Ratos , Triazinas/farmacologia
7.
J Comp Neurol ; 525(17): 3666-3682, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-28758220

RESUMO

Activation of estrogen receptor beta (ERß)-expressing neurons regulates the mammalian stress response via the hypothalamic-pituitary-adrenal (HPA) axis. These neurons densely populate the paraventricular nucleus of the hypothalamus (PVN). Recent research has revealed striking differences between rat and mouse PVN cytochemistry, but careful exploration of PVN ERß neurons in mice has been hindered by a lack of specific ERß antisera. Therefore, we used male and female transgenic mice expressing EGFP under the control of the mouse ERß promoter (ERß-EGFP) to examine the chemical architecture of PVN ERß cells. Using immunohistochemistry, we found that 90% of ERß-immunoreactivity (-ir) colocalized with EGFP. Cellular colocalization of EGFP with neuropeptides, transcription modulators, and neuronal tracers was examined throughout the PVN. ERß-EGFP cells expressed oxytocin more abundantly in the rostral (71 ± 3%) than caudal (33 ± 8%) PVN. Arginine vasopressin colocalized with EGFP more often in females (18 ± 3%) than males (4 ± 1%). Moreover, estrogen receptor α-ir colocalized with ERß-EGFP at low levels (15 ± 3%). Using a corticotropin releasing hormone-cre driver X tdTomato reporter mouse, we found a moderate colocalization with ERß-ir (48 ± 16%) in the middle PVN. Peripheral injection of fluorogold revealed that the rostral PVN ERß-EGFP cells are neuroendocrine neurons whereas non-neuroendocrine (presumably pre-autonomic) ERß-EGFP neurons predominated in the posterior PVN. These data demonstrate chemoarchitectural differences in ERß neurons of the mouse PVN that are different from that previously described for the rat, thus, elucidating potential neuronal pathways involved in the regulation of the HPA axis in mice.


Assuntos
Receptor beta de Estrogênio/metabolismo , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Caracteres Sexuais , Análise de Variância , Animais , Arginina Vasopressina/metabolismo , Hormônio Liberador da Corticotropina/genética , Hormônio Liberador da Corticotropina/metabolismo , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/genética , Feminino , Regulação da Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ocitocina/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
8.
Horm Metab Res ; 49(6): 457-465, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28482370

RESUMO

17ß-Estradiol is known to regulate energy metabolism and body weight. Ovariectomy results in body weight gain while estradiol administration results in a reversal of weight gain. Isoflavones, found in rodent chow, can mimic estrogenic effects making it crucial to understand the role of these compounds on metabolic regulation. The goal of this study is to examine the effect of dietary isoflavones on body weight regulation in the ovariectomized rat. This study will examine how dietary isoflavones can interact with estradiol treatment to affect body weight. Consistent with previous findings, animals fed an isoflavone-rich diet had decreased body weight (p<0.05), abdominal fat (p<0.05), and serum leptin levels (p<0.05) compared to animals fed an isoflavone-free diet. Estradiol replacement resulted in decreased body weight (p<0.05), abdominal fat (p<0.05), and serum leptin (p<0.05). Current literature suggests the involvement of cytokines in the inflammatory response of body weight gain. We screened a host of cytokines and chemokines that may be altered by dietary isoflavones or estradiol replacement. Serum cytokine analysis revealed significant (p<0.05) diet-dependent increases in inflammatory cytokines (keratinocyte-derived chemokine). The isoflavone-free diet in OVX rats resulted in the regulation of the following cytokines and chemokines: interleukin-10, interleukin-18, serum regulated on activation, normal T cell expressed and secreted, and monocyte chemoattractant protein-1 (p<0.05). Overall, these results reveal that estradiol treatment can have differential effects on energy metabolism and body weight regulation depending on the presence of isoflavones in rodent chow.


Assuntos
Peso Corporal/efeitos dos fármacos , Dieta , Estradiol/farmacologia , Terapia de Reposição Hormonal , Isoflavonas/farmacologia , Ovariectomia , Gordura Abdominal/patologia , Adipocinas/sangue , Animais , Citocinas/sangue , Ingestão de Líquidos/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Feminino , Tamanho do Órgão , Ratos Sprague-Dawley , Útero/efeitos dos fármacos , Útero/patologia
9.
Neurosci Lett ; 640: 53-59, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28077306

RESUMO

Phytoestrogens are plant derived, non-steroidal compounds naturally found in rodent chows that potentially have endocrine-disrupting effects. Isoflavones, the most common phytoestrogens, have a similar structure and molecular weight to 17ß-estradiol (E2) and have the ability to bind and activate both isoforms of the estrogen receptor (ER). Most isoflavones have a higher affinity for ERß, which is involved in sexually dimorphic behavioral regulation. The goal of this study was to examine the interaction of isoflavones and E2 presence in the OVX rat on anxiety- and depressive- like behavior and the related BDNF pathophysiology. E2 administration resulted in anxiogenic behaviors when isoflavones were present in the diet (p<0.05), but anxiolytic behaviors when isoflavones were not present (p<0.05). E2 resulted in antidepressive-like behaviors in animals fed an isoflavone-rich diet (p<0.05), with no effect when isoflavones were removed. Increased hippocampal BDNF expression was observed in animals fed an isoflavone-rich diet after E2 administration (p<0.05). BDNF expression in the amygdala and hypothalamus was increased after E2 treatment in animals fed an isoflavone-rich diet. Overall, these results demonstrate that the presence of dietary isoflavones can differentially regulate the effect of E2 replacement on behavior and BDNF expression.


Assuntos
Comportamento Animal/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Encéfalo/efeitos dos fármacos , Estradiol/farmacologia , Interações Alimento-Droga , Isoflavonas/administração & dosagem , Animais , Ansiolíticos/farmacologia , Antidepressivos/farmacologia , Ansiedade/psicologia , Encéfalo/metabolismo , Depressão/psicologia , Dieta , Estradiol/efeitos adversos , Feminino , Aprendizagem em Labirinto/efeitos dos fármacos , Atividade Motora/efeitos dos fármacos , Ovariectomia , Ratos Sprague-Dawley
10.
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.

11.
Birth Defects Res B Dev Reprod Toxicol ; 104(5): 204-17, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26439775

RESUMO

Atrazine (ATZ) was administered daily by gavage to pregnant female Sprague Dawley rats at doses of 0, 6.25, 25 or 50 mg/kg/day, either during gestation, lactation and post-weaning (G/L/PW cohort) to F1 generation female offspring or only from postnatal day (PND 21) until five days after sexual maturation (vaginal opening) when the estrogen-primed, luteinizing hormone (LH) surge was evaluated (PW cohort). Additional subgroups of F1 females received the vehicle or ATZ from PND 21-133 or from PND 120-133. Slight reductions in fertility and the percentage of F1 generation pups surviving to PND 21 in the gestationally exposed 50 mg/kg dose group were accompanied by decreased food intake and body weight of dams and F1 generation offspring. The onset of puberty was delayed in of the F1 generation G/L/PW females at doses of 25 and 50 mg/kg/day. F1 generation females in the PW high-dose ATZ group also experienced a delay in the onset of puberty. ATZ had no effect on peak LH or LH AUC in ovariectomized rats 5 days after sexual maturation, irrespective of whether the F1 generation females were treated from gestation onward or only peripubertally. There was no effect of ATZ treatment on the estrous cycle, peak LH or LH AUC of F1 generation females exposed from gestation through to PND 133 or only for two weeks from PND 120-133. These results indicate that developing females exposed to ATZ are not more sensitive compared to animals exposed to ATZ as young adults.


Assuntos
Envelhecimento/efeitos dos fármacos , Atrazina/toxicidade , Exposição Ambiental , Hormônio Luteinizante/metabolismo , Maturidade Sexual/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Cruzamentos Genéticos , Estradiol/farmacologia , Ciclo Estral/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Feminino , Ratos , Ratos Sprague-Dawley , Análise de Sobrevida , Fatores de Tempo
12.
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
13.
Curr Eye Res ; 40(2): 110-27, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25153712

RESUMO

The actions of gonadal steroid hormones induce morphological sex differences in many tissues in the body, including brain. These occur either during development to organize tissues in a sex-specific pattern and/or in adulthood to activate specific cellular pathways. Cellular and morphological changes in the brain, induced by androgens and estrogens, underlie behavioral sex differences in both reproductive and non-reproductive behaviors, including visual perception. A growing body of evidence indicates that some sex differences related to visual perception arise as the result of the organizational actions of gonadal steroid hormones on cerebral cortical pathways involved in visual processing of objects and movement. This review addresses the influence of gonadal steroids on structural, biochemical and morphological changes in tissues in the brain and body. These effects are extended to consider how gonadal hormone effects may contribute to cognitive sex differences across species that are related to processing within the dorsal and ventral visual streams for motion and objects, respectively. Lastly, this review considers the question of how cognitive sex differences related to processing of movement and objects in humans may be reflective of two types of cognitive style that are only superficially related to gender.


Assuntos
Córtex Cerebral/metabolismo , Cognição/fisiologia , Hormônios Esteroides Gonadais/fisiologia , Receptores Androgênicos/metabolismo , Receptores de Estrogênio/metabolismo , Fatores Sexuais , Percepção Visual/fisiologia , Androgênios/fisiologia , Estrogênios/fisiologia , Feminino , Humanos , Masculino
14.
Physiol Behav ; 129: 287-96, 2014 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-24631553

RESUMO

The hypothalamic-pituitary-adrenal (HPA) axis is activated in response to stressors and is controlled by neurons residing in the paraventricular nucleus of the hypothalamus (PVN). Although gonadal steroid hormones can influence HPA reactivity to stressors, the exact mechanism of action is not fully understood. It is known, however, that estrogen receptor ß (ERß) inhibits HPA reactivity and decreases anxiety-like behavior in rodents. Since ERß is co-expressed with oxytocin (OT) in neurons of the PVN, an ERß-selective agonist was utilized to test the whether ERß decreases stress-induced HPA reactivity and anxiety-like behaviors via an OTergic pathway. Adult gonadectomized male and female rats were administered diarylpropionitrile, or vehicle, peripherally for 5days. When tested for anxiety-like behavior on the elevated plus maze (EPM), diarylpropionitrile-treated males and females significantly increased time on the open arm of the EPM compared to vehicle controls indicating that ERß reduces anxiety-like behaviors. One week after behavioral evaluation, rats were subjected to a 20minute restraint stress. Treatment with diarylpropionitrile reduced CORT and ACTH responses in both males and females. Subsequently, another group of animals was implanted with cannulae directed at the lateral ventricle. One week later, rats underwent the same protocol as above but with the additional treatment of intracerebroventricular infusion with an OT antagonist (des Gly-NH2 d(CH2)5 [Tyr(Me)(2), Thr(4)] OVT) or VEH, 20min prior to behavioral evaluation. OT antagonist treatment blocked the effects of diarylpropionitrile on the display of anxiety-like behaviors and plasma CORT levels. These data indicate that ERß and OT interact to modulate the HPA reactivity and the display of anxiety-like behaviors.


Assuntos
Ansiedade/metabolismo , Receptor beta de Estrogênio/metabolismo , Ocitocina/metabolismo , Estresse Psicológico/metabolismo , Hormônio Adrenocorticotrópico/sangue , Animais , Ansiedade/induzido quimicamente , Ansiedade/tratamento farmacológico , Fármacos do Sistema Nervoso Central/farmacologia , Corticosterona/sangue , Transtorno Depressivo/induzido quimicamente , Transtorno Depressivo/tratamento farmacológico , Transtorno Depressivo/metabolismo , Receptor beta de Estrogênio/agonistas , Comportamento Exploratório/efeitos dos fármacos , Comportamento Exploratório/fisiologia , Feminino , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Nitrilas/farmacologia , Ornipressina/análogos & derivados , Ornipressina/farmacologia , Ocitocina/antagonistas & inibidores , Propionatos/farmacologia , Ratos Sprague-Dawley , Restrição Física , Fatores Sexuais , Estresse Psicológico/induzido quimicamente , Estresse Psicológico/tratamento farmacológico
15.
Front Neuroendocrinol ; 35(2): 197-220, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24246855

RESUMO

The hypothalamo-pituitary-adrenal (HPA) axis represents a complex neuroendocrine feedback loop controlling the secretion of adrenal glucocorticoid hormones. Central to its function is the paraventricular nucleus of the hypothalamus (PVN) where neurons expressing corticotropin releasing factor reside. These HPA motor neurons are a primary site of integration leading to graded endocrine responses to physical and psychological stressors. An important regulatory factor that must be considered, prior to generating an appropriate response is the animal's reproductive status. Thus, PVN neurons express androgen and estrogen receptors and receive input from sites that also express these receptors. Consequently, changes in reproduction and gonadal steroid levels modulate the stress response and this underlies sex differences in HPA axis function. This review examines the make up of the HPA axis and hypothalamo-pituitary-gonadal (HPG) axis and the interactions between the two that should be considered when exploring normal and pathological responses to environmental stressors.


Assuntos
Hormônio Liberador da Corticotropina/fisiologia , Hormônios Esteroides Gonadais/fisiologia , Sistema Hipotálamo-Hipofisário/fisiologia , Sistema Hipófise-Suprarrenal/fisiologia , Estresse Fisiológico/fisiologia , Animais , Humanos , Receptores de Estrogênio/fisiologia
16.
J Comp Neurol ; 522(2): 358-71, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23818057

RESUMO

Estrogen receptors regulate multiple brain functions, including stress, sexual, and memory-associated behaviors as well as controlling neuroendocrine and autonomic function. During development, estrogen signaling is involved in programming adult sex differences in physiology and behavior. Expression of estrogen receptor α changes across development in a region-specific fashion. By contrast, estrogen receptor ß (ERß) is expressed in many brain regions, yet few studies have explored sex and developmental differences in its expression, largely because of the absence of selective reagents for anatomical localization of the protein. This study utilized bacterial artificial chromosome transgenic mice expressing ERß identified by enhanced green fluorescent protein (EGFP) to compare expression levels and distribution of ERß in the male and female mouse forebrain on the day of birth (P0), on postnatal day 4 (P4), and on P21. By using qualitative analysis, we mapped the distribution of ERß-EGFP and found developmental alterations in ERß expression within the cortex, hippocampus, and hypothalamic regions including the arcuate, ventromedial, and paraventricular nuclei. We also report a sex difference in ERß in the bed nucleus of the stria terminalis, with males showing greater expression at P4 and P21. Another sex difference was found in the anteroventral periventricular nucleus of P21, but not P0 or P4, mice, in which ERß-EGFP-immunoreactive cells were densely clustered near the third ventricle in females but not males. These developmental changes and sex differences in ERß indicate a mechanism through which estrogens might differentially affect brain functions or program adult physiology at select times during development.


Assuntos
Receptor beta de Estrogênio/biossíntese , Prosencéfalo/crescimento & desenvolvimento , Prosencéfalo/metabolismo , Caracteres Sexuais , Fatores Etários , Animais , Cromossomos Artificiais Bacterianos , Feminino , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Transgênicos
17.
J Neurochem ; 127(4): 487-95, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24033289

RESUMO

In the dorsal raphe nucleus, 17ß-estradiol (E2) increases the expression of the brain-specific, rate-limiting enzyme for serotonin biosynthesis, tryptophan hydroxylase-2 (Tph2). Although estrogen receptor beta (ERß) has been localized to Tph2 neurons, little is known about the transcriptional regulation of the Tph2 gene by estrogen. Since the ERß agonist, diarylpropionitrile (DPN) also increases Tph2 expression, we tested the hypothesis that E2 regulates the Tph2 promoter through direct interactions with ERß. A serotonergic cell line, B14, which endogenously expresses ERß was transiently transfected with a fragment of the human TPH2 5'-untranslated region (5'-UTR) cloned into a luciferase reporter vector (TPH2-luc). Treatment with E2 or DPN caused a dose-dependent increase of TPH2-luc activity. In contrast, E2 conjugated to bovine serum albumin, which is cell membrane impermeable, had no effect on TPH2-luc activity. An estrogen receptor (ER) antagonist blocked E2 or DPN-induced TPH2-luc activity suggesting a classical ER mechanism. In silico analysis revealed an estrogen-response element (ERE) half-site located within the TPH2 5'-UTR. Deletion and site-directed mutation of this site abolished ligand-induced TPH2-luc activity. These results support the concept that there is a direct and functional interaction between E2:ERß and the ERE half-site of the TPH2 promoter to regulate Tph2 expression. We illustrate a direct regulation of the TPH2 transcription by estradiol and ERß via a newly identified ERE half-site within the TPH2 promoter: (i) Estradiol- or an ERß agonist-induced TPH2 transcription was blocked by an ER antagonist, while (ii) membrane impermeable form of estradiol did not induce transcription. (iii) Deletion or mutation of the ERE half-site abolished ligand-induced TPH2 transcription.


Assuntos
Estradiol/metabolismo , Receptor beta de Estrogênio/metabolismo , Estrogênios/metabolismo , Elementos de Resposta , Triptofano Hidroxilase/metabolismo , Regiões 5' não Traduzidas , Animais , Linhagem Celular , Estradiol/farmacologia , Receptor beta de Estrogênio/agonistas , Estrogênios/farmacologia , Humanos , Camundongos , Dados de Sequência Molecular , Nitrilas/farmacologia , Regiões Promotoras Genéticas , Propionatos/farmacologia , Ratos , Transdução de Sinais , Transcrição Gênica , Triptofano Hidroxilase/genética
18.
Brain Res ; 1529: 74-82, 2013 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-23880372

RESUMO

In rodents, the hypothalamo-pituitary-adrenal (HPA) axis is controlled by a precise regulatory mechanism that is influenced by circulating gonadal and adrenal hormones. In males, gonadectomy increases the adrenocorticotropic hormone (ACTH) and corticosterone (CORT) response to stressors, and androgen replacement returns the response to that of the intact male. Testosterone (T) actions in regulating HPA activity may be through aromatization to estradiol, or by 5α-reduction to the more potent androgen, dihydrotestosterone (DHT). To determine if the latter pathway is involved, we assessed the function of the HPA axis response to restraint stress following hormone treatments, or after peripheral or central treatment with the 5α-reductase inhibitor, finasteride. Initially, we examined the timecourse whereby gonadectomy alters the CORT response to restraint stress. Enhanced CORT responses were evident within 48 h following gonadectomy. Correspondingly, treatment of intact male rats with the 5α-reductase inhibitor, finasteride, for 48 h, enhanced the CORT and ACTH response to restraint stress. Peripheral injections of gonadectomized male rats with DHT or T for 48 h reduced the ACTH and CORT response to restraint stress. The effects of T, but not DHT, could be blocked by the third ventricle administration of finasteride prior to stress application. These data indicate that the actions of T in modulating HPA axis activity involve 5α-reductase within the central nervous system. These results further our understanding of how T acts to modulate the neuroendocrine stress responses and indicate that 5α reduction to DHT is a necessary step for T action.


Assuntos
Inibidores de 5-alfa Redutase/farmacologia , Finasterida/farmacologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Estresse Fisiológico/fisiologia , Testosterona/metabolismo , Hormônio Adrenocorticotrópico/metabolismo , Animais , Corticosterona/metabolismo , Di-Hidrotestosterona/farmacologia , Modelos Animais de Doenças , Estradiol/farmacologia , Sistema Hipotálamo-Hipofisário/metabolismo , Injeções Intra-Articulares , Masculino , Orquiectomia , Sistema Hipófise-Suprarrenal/metabolismo , Radioimunoensaio , Ratos , Ratos Sprague-Dawley , Restrição Física/efeitos adversos , Testosterona/sangue , Fatores de Tempo
19.
Endocrinology ; 154(5): 1802-12, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23515287

RESUMO

Testosterone has been shown to suppress the acute stress-induced activation of the hypothalamic-pituitary-adrenal axis; however, the mechanisms underlying this response remain unclear. The hypothalamic-pituitary-adrenal axis is regulated by a neuroendocrine subpopulation of medial parvocellular neurons in the paraventricular nucleus of the hypothalamus (PVN). These neurons are devoid of androgen receptors (ARs). Therefore, a possibility is that the PVN target neurons respond to a metabolite in the testosterone catabolic pathway via an AR-independent mechanism. The dihydrotestosterone metabolite, 5α-androstane-3ß,17ß-diol (3ß-diol), binds and activates estrogen receptor-ß (ER-ß), the predominant ER in the PVN. In the PVN, ER-ß is coexpressed with oxytocin (OT). Therefore, we tested the hypothesis that 3ß-diol regulates OT expression through ER-ß activation. Treatment of ovariectomized rats with estradiol benzoate or 3ß-diol for 4 days increased OT mRNA selectively in the midcaudal, but not rostral PVN compared with vehicle-treated controls. 3ß-Diol treatment also increased OT mRNA in the hypothalamic N38 cell line in vitro. The functional interactions between 3ß-diol and ER-ß with the human OT promoter were examined using an OT promoter-luciferase reporter construct (OT-luc). In a dose-dependent manner, 3ß-diol treatment increased OT-luc activity when cells were cotransfected with ER-ß, but not ER-α. The 3ß-diol-induced OT-luc activity was reduced by deletion of the promoter region containing the composite hormone response element (cHRE). Point mutations of the cHRE also prevented OT-luc activation by 3ß-diol. These results indicate that 3ß-diol induces OT promoter activity via ER-ß-cHRE interactions.


Assuntos
Androstano-3,17-diol/farmacologia , Receptor beta de Estrogênio/fisiologia , Ocitocina/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos , Androgênios/metabolismo , Androstano-3,17-diol/metabolismo , Animais , Células Cultivadas , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Feminino , Humanos , Camundongos , Ovariectomia , Ocitocina/metabolismo , Regiões Promotoras Genéticas/fisiologia , Ratos , Ratos Sprague-Dawley , Elementos de Resposta/efeitos dos fármacos , Elementos de Resposta/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Ativação Transcricional/genética
20.
Am J Pathol ; 182(2): 485-504, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23332367

RESUMO

Duchenne muscular dystrophy (DMD) is a severe disorder characterized by progressive muscle wasting,respiratory and cardiac impairments, and premature death. No treatment exists so far, and the identification of active substances to fight DMD is urgently needed. We found that tamoxifen, a drug used to treat estrogen-dependent breast cancer, caused remarkable improvements of muscle force and of diaphragm and cardiac structure in the mdx(5Cv) mouse model of DMD. Oral tamoxifen treatment from 3 weeks of age for 15 months at a dose of 10 mg/kg/day stabilized myofiber membranes, normalized whole body force, and increased force production and resistance to repeated contractions of the triceps muscle above normal values. Tamoxifen improved the structure of leg muscles and diminished cardiac fibrosis by~ 50%. Tamoxifen also reduced fibrosis in the diaphragm, while increasing its thickness,myofiber count, and myofiber diameter, thereby augmenting by 72% the amount of contractile tissue available for respiratory function. Tamoxifen conferred a markedly slower phenotype to the muscles.Tamoxifen and its metabolites were present in nanomolar concentrations in plasma and muscles,suggesting signaling through high-affinity targets. Interestingly, the estrogen receptors ERa and ERb were several times more abundant in dystrophic than in normal muscles, and tamoxifen normalized the relative abundance of ERb isoforms. Our findings suggest that tamoxifen might be a useful therapy for DMD.


Assuntos
Antineoplásicos/uso terapêutico , Distrofia Muscular Animal/tratamento farmacológico , Distrofia Muscular Animal/patologia , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/patologia , Tamoxifeno/uso terapêutico , Animais , Antineoplásicos/farmacologia , Comportamento Animal/efeitos dos fármacos , Biomarcadores/metabolismo , Fenômenos Biomecânicos/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Creatina Quinase/sangue , Diafragma/patologia , Diafragma/fisiopatologia , Modelos Animais de Doenças , Comportamento Alimentar/efeitos dos fármacos , Fibrose , Camundongos , Contração Muscular/efeitos dos fármacos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/patologia , Distrofia Muscular Animal/sangue , Distrofia Muscular Animal/fisiopatologia , Distrofia Muscular de Duchenne/sangue , Distrofia Muscular de Duchenne/fisiopatologia , Miocárdio/patologia , Tamanho do Órgão/efeitos dos fármacos , Receptores de Estrogênio/metabolismo , Tamoxifeno/sangue , Tamoxifeno/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA