Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Horm Behav ; 164: 105594, 2024 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-38917776

RESUMO

Menopause is an endocrine shift leading to increased vulnerability for cognitive impairment and dementia risk factors, in part due to loss of neuroprotective circulating estrogens. Systemic replacement of estrogen post-menopause has limitations, including risk for estrogen-sensitive cancers. A promising therapeutic approach therefore might be to deliver estrogen only to the brain. We examined whether we could enhance cognitive performance by delivering estrogen exclusively to the brain in ovariectomized mice (a surgical menopause model). We treated mice with the prodrug 10ß,17ß-dihydroxyestra-1,4-dien-3-one (DHED), which can be administered systemically but is converted to 17ß-estradiol only in the brain. Young and middle-aged C57BL/6 J mice received ovariectomy and subcutaneous implant containing vehicle or DHED and underwent cognitive testing to assess memory after 1-3.5 months of treatment. Low and medium doses of DHED did not alter metabolic status in middle-aged mice. In both age groups, DHED treatment improved spatial memory in ovariectomized mice. Additional testing in middle-aged mice showed that DHED treatment improved working and recognition memory in ovariectomized mice. These results lay the foundation for future studies determining if this intervention is as efficacious in models of dementia with comorbid risk factors.

2.
Horm Behav ; 162: 105528, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38503191

RESUMO

Testosterone is a powerful steroid hormone that can impact the brain and behavior in various ways, including regulating behavioral and neuroendocrine (hypothalamic-pituitary-adrenal (HPA) axis) stress responses. Early in life androgens can act to alter development of brain regions associated with stress regulation, which ultimately impacts the display of stress responses later in life. Adult circulating androgens can also influence the expression of distinct genes and proteins that regulate stress responses. These changes in the brain are hypothesized to underlie the potent effects of androgens in regulating behaviors related to stress and stress-induced activation of the HPA axis. Androgens can induce alterations in these functions through direct binding to the androgen receptor (AR) or following conversion to estrogens and subsequent binding to estrogen receptors including estrogen receptor alpha (ERα), beta (ERß), and G protein-coupled estrogen receptor 1 (GPER1). In this review, we focus on the role of androgens in regulating behavioral and neuroendocrine stress responses at different stages of the lifespan and the sex hormone receptors involved in regulating these effects. We also review the specific brain regions and cell phenotypes upon which androgens are proposed to act to regulate stress responses with an emphasis on hypothalamic and extended amygdala subregions. This knowledge of androgen effects on these neural systems is critical for understanding how sex hormones regulate stress responses.


Assuntos
Androgênios , Sistema Hipotálamo-Hipofisário , Sistema Hipófise-Suprarrenal , Estresse Psicológico , Animais , Humanos , Androgênios/fisiologia , Androgênios/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Receptores Androgênicos/metabolismo , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia
3.
bioRxiv ; 2023 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-37609180

RESUMO

Menopause is a major endocrinological shift that leads to an increased vulnerability to the risk factors for cognitive impairment and dementia. This is thought to be due to the loss of circulating estrogens, which exert many potent neuroprotective effects in the brain. Systemic replacement of estrogen post-menopause has many limitations, including increased risk for estrogen-sensitive cancers. A more promising therapeutic approach therefore might be to deliver estrogen only to the brain thus limiting adverse peripheral side effects. We examined whether we could enhance cognitive performance by delivering estrogen exclusively to the brain in post-menopausal mice. We modeled surgical menopause via bilateral ovariectomy (OVX). We treated mice with the pro-drug 10ß,17ß-dihydroxyestra-1,4-dien-3-one (DHED), which can be administered systemically but is converted to 17ß-estradiol only in the brain. Young (2.5-month) and middle-aged (11-month-old) female C57BL/6J mice received ovariectomy and a subcutaneous implant containing vehicle (cholesterol) or DHED. At 3.5 months old (young group) and 14.5 months old (middle-aged group), mice underwent behavior testing to assess memory. DHED did not significantly alter metabolic status in middle-aged, post-menopausal mice. In both young and middle-aged mice, the brain-specific estrogen DHED improved spatial memory. Additional testing in middle-aged mice also showed that DHED improved working and recognition memory. These promising results lay the foundation for future studies aimed at determining if this intervention is as efficacious in models of dementia that have comorbid risk factors.

4.
Front Physiol ; 14: 1215535, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37440997

RESUMO

Introduction: The response of the brain to space radiation is an important concern for astronauts during space missions. Therefore, we assessed the response of the brain to 28Si ion irradiation (600 MeV/n), a heavy ion present in the space environment, on cognitive performance and whether the response is associated with altered DNA methylation in the hippocampus, a brain area important for cognitive performance. Methods: We determined the effects of 28Si ion irradiation on object recognition, 6-month-old mice irradiated with 28Si ions (600 MeV/n, 0.3, 0.6, and 0.9 Gy) and cognitively tested two weeks later. In addition, we determined if those effects were associated with alterations in hippocampal networks and/or hippocampal DNA methylation. Results: At 0.3 Gy, but not at 0.6 Gy or 0.9 Gy, 28Si ion irradiation impaired cognition that correlated with altered gene expression and 5 hmC profiles that mapped to specific gene ontology pathways. Comparing hippocampal DNA hydroxymethylation following proton, 56Fe ion, and 28Si ion irradiation revealed a general space radiation synaptic signature with 45 genes that are associated with profound phenotypes. The most significant categories were glutamatergic synapse and postsynaptic density. Discussion: The brain's response to space irradiation involves novel excitatory synapse and postsynaptic remodeling.

5.
PLoS One ; 18(3): e0282293, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36862730

RESUMO

It is widely established that gonadal hormones are fundamental to modulating and organizing the sex-specific nature of reproductive behaviors. Recently we proposed that context fear conditioning (CFC) may emerge in a sex-specific manner organized prior to the pubertal surge of gonadal hormones. Here we sought to determine the necessity of male and female gonadal hormones secreted at critical periods of development upon context fear learning. We tested the organizational hypothesis that neonatal and pubertal gonadal hormones play a permanent role in organizing contextual fear learning. We demonstrate that the postnatal absence of gonadal hormones by neonatal orchiectomy (oRX) in males and ovariectomy (oVX) in females resulted in an attenuation of CFC in adult males and an enhancement of CFC in adult females. In females, the gradual introduction of estrogen before conditioning partially rescued this effect. However, the decrease of CFC in adult males was not rescued by introducing testosterone before conditioning. Next, at a further point in development, preventing the pubertal surge of gonadal hormones by prepubertal oRX in males resulted in a reduction in adult CFC. In contrast, in females, prepubertal oVX did not alter adult CFC. However, the adult introduction of estrogen in prepubertal oVX rats reduced adult CFC. Lastly, the adult-specific deletion of gonadal hormones by adult oRX or oVX alone or replacement of testosterone or estrogen did not alter CFC. Consistent with our hypothesis, we provide initial evidence that gonadal hormones at early periods of development exert a vital role in the organization and development of CFC in male and female rats.


Assuntos
Hormônios Gonadais , Aprendizagem , Feminino , Masculino , Animais , Ratos , Estrogênios , Medo , Testosterona
6.
Horm Behav ; 156: 105448, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38344954

RESUMO

Gonadal hormone actions through androgen receptor (AR) and estrogen receptor alpha (ERα) regulate sex differences in hypothalamic-pituitary-adrenal (HPA) axis responsivity and stress-related behaviors. Here we tested whether corticotropin releasing factor (CRF) expressing neurons, which are widely known to regulate neuroendocrine and behavioral stress responses, co-express AR and ERα as a potential mechanism for gonadal hormone regulation of these responses. Using Crh-IRES-Cre::Ai9 reporter mice we report high co-localization of AR in CRF neurons within the medial preoptic area (MPOA), bed nucleus of the stria terminalis (BST), medial amygdala (MeA), and ventromedial hypothalamus (VMH), moderate levels within the central amygdala (CeA) and low levels in the paraventricular hypothalamus (PVN). Sex differences in CRF/AR co-expression were found in the principal nucleus of the BST (BSTmpl), CeA, MeA, and VMH (males>females). CRF co-localization with ERα was generally lower relative to AR co-localization. However, high co-expression was found within the MPOA, AVPV, and VMH, with moderate co-expression in the arcuate nucleus (ARC), BST, and MeA and low levels in the PVN and CeA. Sex differences in CRF/ERα co-localization were found in the BSTmpl and PVN (males>females). Finally, we assessed neural activation of CRF neurons in restraint-stressed mice and found greater CRF/c-Fos co-expression in females in the BSTmpl and periaqueductal gray, while co-expression was higher in males within the ARC and dorsal CA1. Given the known role of CRF in regulating behavioral stress responses and the HPA axis, AR/ERα co-expression and sex-specific activation of CRF cell groups indicate potential mechanisms for modulating sex differences in these functions.


Assuntos
Hormônio Liberador da Corticotropina , Receptor alfa de Estrogênio , Camundongos , Feminino , Masculino , Animais , Hormônio Liberador da Corticotropina/genética , Hormônio Liberador da Corticotropina/metabolismo , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Caracteres Sexuais , Receptores Androgênicos/genética , Sistema Hipófise-Suprarrenal/metabolismo , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Proteínas Proto-Oncogênicas c-fos , Neurônios/metabolismo , Hormônios Gonadais , Núcleo Hipotalâmico Paraventricular/metabolismo
7.
Endocrinology ; 161(11)2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32798220

RESUMO

GnRH neurons are central regulators of reproduction and respond to factors affecting fertility, such as stress. Corticotropin-releasing hormone (CRH) is released during stress response. In brain slices from unstressed controls, CRH has opposite, estradiol-dependent effects on GnRH neuron firing depending on the CRH receptor activated; activating CRHR-1 stimulates whereas activating CRHR-2 suppresses activity. We investigated possible direct and indirect mechanisms. Mice were ovariectomized and either not treated further (OVX) or given a capsule producing high positive feedback (OVX + E) or low negative feedback (OVX + low E) physiologic circulating estradiol levels. We tested possible direct effects on GnRH neurons by altering voltage-gated potassium currents. Two types of voltage-gated potassium currents (transient IA and sustained IK) were measured; neither CRHR-1 nor CRHR-2 agonists altered potassium current density in GnRH neurons from OVX + E mice. Further, neither CRH nor receptor-specific agonists altered action potential generation in response to current injection in GnRH neurons from OVX + E mice. To test the possible indirect actions, GABAergic postsynaptic currents were monitored. A CRHR-1 agonist increased GABAergic transmission frequency to GnRH neurons from OVX + E, but not OVX, mice, whereas a CRHR-2 agonist had no effect. Finally, we tested if CRH alters the firing rate of arcuate kisspeptin neurons, which provide an important excitatory neuromodulatory input to GnRH neurons. CRH did not acutely alter firing activity of these neurons from OVX, OVX + E or OVX + low E mice. These results suggest CRH increases GnRH neuron activity in an estradiol-dependent manner in part by activating GABAergic afferents. Mechanisms underlying inhibitory effects of CRH remain unknown.


Assuntos
Hormônio Liberador da Corticotropina/análogos & derivados , Estradiol/farmacologia , Neurônios/efeitos dos fármacos , Peptídeos Cíclicos/farmacologia , Transmissão Sináptica/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Animais , Hormônio Liberador da Corticotropina/farmacologia , Estradiol/sangue , Retroalimentação Fisiológica/efeitos dos fármacos , Retroalimentação Fisiológica/fisiologia , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Neurônios/fisiologia , Ovariectomia , Receptores de Hormônio Liberador da Corticotropina/agonistas , Urocortinas/farmacologia , Ácido gama-Aminobutírico/metabolismo
8.
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
9.
Neuroscience ; 409: 195-203, 2019 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-31055007

RESUMO

Sex differences in neural structures are generally believed to underlie sex differences reported in anxiety, depression, and the hypothalamic-pituitary-adrenal axis, although the specific circuitry involved is largely unclear. Using a corticotropin-releasing factor receptor 1 (CRFR1) reporter mouse line, we report a sexually dimorphic distribution of CRFR1 expressing cells within the paraventricular hypothalamus (PVN; males > females). Relative to adult levels, PVN CRFR1-expressing cells are sparse and not sexually dimorphic at postnatal days 0, 4, or 21. This suggests that PVN cells might recruit CRFR1 during puberty or early adulthood in a sex-specific manner. The adult sex difference in PVN CRFR1 persists in old mice (20-24 months). Adult gonadectomy (6 weeks) resulted in a significant decrease in CRFR1-immunoreactive cells in the male but not female PVN. CRFR1 cells show moderate co-expression with estrogen receptor alpha (ERα) and high co-expression with androgen receptor, indicating potential mechanisms through which circulating gonadal hormones might regulate CRFR1 expression and function. Finally, we demonstrate that a psychological stressor, restraint stress, induces a sexually dimorphic pattern of neural activation in PVN CRFR1 cells (males >females) as assessed by co-localization with the transcription/neural activation marker phosphorylated CREB. Given the known role of CRFR1 in regulating stress-associated behaviors and hormonal responses, this CRFR1 PVN sex difference might contribute to sex differences in these functions.


Assuntos
Sistema Hipotálamo-Hipofisário/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Caracteres Sexuais , Estresse Psicológico/metabolismo , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Receptor alfa de Estrogênio/metabolismo , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Fosforilação , Restrição Física
10.
Behav Brain Res ; 366: 67-76, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-30902659

RESUMO

Chronic methamphetamine (MA) use can lead to increased symptoms of depression and anxiety during abstinence. Less is known about the specific brain regions that are altered following repeated MA that may be associated with these behavioral perturbations. Furthermore, MA has been reported to recruit and activate microglia in the brain, which may exacerbate stress-associated behavioral changes. In the present study, male and female mice were injected with MA (5 mg/kg) or saline once daily for 10 days, and during early withdrawal were assessed for alterations in immediate early gene (c-Fos) responses to a forced swim stressor. Chronic MA exposure increased floating and decreased swim time in the forced swim test in male and female mice tested 48 h after the final dose, indicating elevated depressive-like behavior. Furthermore, assessment of nest building, a measure of distress or despair-like behavior, revealed a sex-specific effect with only MA-treated females showing impairments. The c-Fos response to forced swim was attenuated by prior MA exposure in the central amygdala, CA3 hippocampal region, prefrontal cortex, and bed nucleus of the stria terminalis (BST). In the BST this attenuation occurred only in males. Neither the total number of microglia or activated microglia were altered by chronic MA exposure in regions examined. The primary findings indicate that chronic MA exposure attenuates activation of select stress-associated brain regions, a dysregulation that might contribute to alterations in mood-related behaviors.


Assuntos
Metanfetamina/metabolismo , Neurônios/metabolismo , Síndrome de Abstinência a Substâncias/metabolismo , Animais , Ansiedade/metabolismo , Encéfalo/metabolismo , Região CA3 Hipocampal/metabolismo , Estimulantes do Sistema Nervoso Central/farmacologia , Corticosterona/farmacologia , Depressão/metabolismo , Teste de Esforço/métodos , Feminino , Sistema Hipotálamo-Hipofisário/metabolismo , Masculino , Metanfetamina/efeitos adversos , Metanfetamina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Estresse Fisiológico/fisiologia , Síndrome de Abstinência a Substâncias/fisiopatologia , Natação
11.
J Comp Neurol ; 527(6): 1056-1069, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30499109

RESUMO

Corticotropin-releasing factor binds with high affinity to CRF receptor 1 (CRFR1) and is implicated in stress-related mood disorders such as anxiety and depression. Using a validated CRFR1-green fluorescent protein (GFP) reporter mouse, our laboratory recently discovered a nucleus of CRFR1 expressing cells that is prominent in the female rostral anteroventral periventricular nucleus (AVPV/PeN), but largely absent in males. This sex difference is present in the early postnatal period and remains dimorphic into adulthood. The present investigation sought to characterize the chemical composition and gonadal hormone regulation of these sexually dimorphic CRFR1 cells using immunohistochemical procedures. We report that CRFR1-GFP-ir cells within the female AVPV/PeN are largely distinct from other dimorphic cell populations (kisspeptin, tyrosine hydroxylase). However, CRFR1-GFP-ir cells within the AVPV/PeN highly co-express estrogen receptor alpha as well as glucocorticoid receptor. A single injection of testosterone propionate or estradiol benzoate on the day of birth completely eliminates the AVPV/PeN sex difference, whereas adult gonadectomy has no effect on CRFR1-GFP cell number. These results indicate that the AVPV/PeN CRFR1 is regulated by perinatal but not adult gonadal hormones. Finally, female AVPV/PeN CRFR1-GFP-ir cells are activated following an acute 30-min restraint stress, as assessed by co-localization of CRFR1-GFP cells with phosphorylated (p) CREB. CRFR1-GFP/pCREB cells were largely absent in the male AVPV/PeN. Together, these data indicate a stress and gonadal hormone responsive nucleus that is unique to females and may contribute to sex-specific stress responses.


Assuntos
Hipotálamo Anterior/citologia , Neurônios/citologia , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Caracteres Sexuais , Animais , Feminino , Hormônios Gonadais/fisiologia , Hipotálamo Anterior/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo
12.
Brain Res ; 1677: 74-85, 2017 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-28941573

RESUMO

Sex differences in patterns of methamphetamine (MA) abuse have been reported with females (humans and rodents) showing an elevated addiction phenotype. Previous findings indicate MA-induced hypothalamic-pituitary-adrenal (HPA) axis activation is also sexually dimorphic with females exhibiting an elevated glucocorticoid release and differential neural activation patterns within HPA axis-associated brain regions. These effects may contribute to sex differences in abuse. To determine the role of gonadal hormones in mediating sex differences in MA-induced glucocorticoids, male and female C57BL/6J mice were gonadectomized or sham-operated, and following recovery, injected with MA (5mg/kg) and sacrificed 60min or 120min later. Blood was collected for corticosterone radioimmunoassay, and brains were used to assess c-Fos, and c-Fos co-localization with glucocorticoid receptor (GR). At 120min after MA injection, corticosterone levels were elevated in females compared to males and gonadectomy in males increased corticosterone to female levels. C-Fos was greater in females than males in the medial preoptic area, bed nucleus of the stria terminalis, basolateral amygdala, and central amygdala. Female gonadectomy had little effect on either corticosterone or c-Fos, while male gonadectomy elevated c-Fos in the central amygdala. Relative to sham males, gonadectomized males also showed decreased c-Fos/GR cell number in the CA3 hippocampal area compared to sham males, indicating a central site for attenuated negative feedback. Together, these findings indicate that androgens regulate MA-induced activation of the HPA axis, potentially by enhancing negative feedback. These sex and gonadal hormone effects on the HPA axis may contribute to sex differences in MA abuse patterns.


Assuntos
Estimulantes do Sistema Nervoso Central/farmacologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Metanfetamina/farmacologia , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Caracteres Sexuais , Transtornos Relacionados ao Uso de Anfetaminas/metabolismo , Animais , Temperatura Corporal/efeitos dos fármacos , Temperatura Corporal/fisiologia , Corticosterona/sangue , Feminino , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipotálamo-Hipofisário/patologia , Imuno-Histoquímica , Masculino , Camundongos Endogâmicos C57BL , Orquiectomia , Ovariectomia , Sistema Hipófise-Suprarrenal/metabolismo , Sistema Hipófise-Suprarrenal/patologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores de Glucocorticoides/metabolismo
13.
Sci Rep ; 7(1): 10227, 2017 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-28860502

RESUMO

The brain's response to radiation exposure is an important concern for patients undergoing cancer therapy and astronauts on long missions in deep space. We assessed whether this response is specific and prolonged and is linked to epigenetic mechanisms. We focused on the response of the hippocampus at early (2-weeks) and late (20-week) time points following whole body proton irradiation. We examined two forms of DNA methylation, cytosine methylation (5mC) and hydroxymethylation (5hmC). Impairments in object recognition, spatial memory retention, and network stability following proton irradiation were observed at the two-week time point and correlated with altered gene expression and 5hmC profiles that mapped to specific gene ontology pathways. Significant overlap was observed between DNA methylation changes at the 2 and 20-week time points demonstrating specificity and retention of changes in response to radiation. Moreover, a novel class of DNA methylation change was observed following an environmental challenge (i.e. space irradiation), characterized by both increased and decreased 5hmC levels along the entire gene body. These changes were mapped to genes encoding neuronal functions including postsynaptic gene ontology categories. Thus, the brain's response to proton irradiation is both specific and prolonged and involves novel remodeling of non-random regions of the epigenome.


Assuntos
Metilação de DNA/efeitos da radiação , Epigenômica/métodos , Hipocampo/efeitos da radiação , Irradiação Corporal Total/métodos , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/análise , 5-Metilcitosina/efeitos da radiação , Animais , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos da radiação , Redes Reguladoras de Genes/efeitos da radiação , Hipocampo/química , Masculino , Aprendizagem em Labirinto/efeitos da radiação , Camundongos , Prótons/efeitos adversos , Análise de Sequência de RNA , Aprendizagem Espacial/efeitos da radiação , Fatores de Tempo
14.
EBioMedicine ; 3: 26-42, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26870815

RESUMO

Obesity, metabolic syndrome (MetS) and type 2 diabetes (T2D) are associated with decreased cognitive function. While weight loss and T2D remission result in improvements in metabolism and vascular function, it is less clear if these benefits extend to cognitive performance. Here, we highlight the malleable nature of MetS-associated cognitive dysfunction using a mouse model of high fat diet (HFD)-induced MetS. While learning and memory was generally unaffected in mice with type 1 diabetes (T1D), multiple cognitive impairments were associated with MetS, including deficits in novel object recognition, cued fear memory, and spatial learning and memory. However, a brief reduction in dietary fat content in chronic HFD-fed mice led to a complete rescue of cognitive function. Cerebral blood volume (CBV), a measure of vascular perfusion, was decreased during MetS, was associated with long term memory, and recovered following the intervention. Finally, repeated infusion of plasma collected from age-matched, low fat diet-fed mice improved memory in HFD mice, and was associated with a distinct metabolic profile. Thus, the cognitive dysfunction accompanying MetS appears to be amenable to treatment, related to cerebrovascular function, and mitigated by systemic factors.


Assuntos
Transtornos Cognitivos/etiologia , Transtornos Cognitivos/metabolismo , Gorduras na Dieta/metabolismo , Síndrome Metabólica/complicações , Síndrome Metabólica/metabolismo , Animais , Comportamento Animal , Circulação Cerebrovascular , Análise por Conglomerados , Dieta Hiperlipídica , Modelos Animais de Doenças , Feminino , Aprendizagem em Labirinto , Síndrome Metabólica/fisiopatologia , Metaboloma , Metabolômica/métodos , Camundongos , Obesidade/metabolismo , Reconhecimento Psicológico , Redução de Peso
15.
Psychopharmacology (Berl) ; 233(3): 381-92, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26525566

RESUMO

RATIONALE: Chronic methamphetamine (MA) abuse leads to dependence and symptoms of withdrawal after use has ceased. Negative mood states associated with withdrawal, as well as drug reinstatement, have been linked to drug-induced disruption of the hypothalamic-pituitary-adrenal (HPA) axis. However, effects of chronic MA exposure or acute MA exposure following withdrawal on neural activation patterns within brain regions that regulate the HPA axis are unknown. OBJECTIVES: In this study, neural activation patterns were assessed by quantification of c-Fos protein in mice exposed to different regimens of MA administration. METHODS: (Experiment 1) Adult male mice were treated with MA (5 mg/kg) or saline once or once daily for 10 days. (Experiment 2) Mice were treated with MA or saline once daily for 10 days and following a 10-day withdrawal period were re-administered a final dose of MA or saline. c-Fos was quantified in brains after the final injection. RESULTS: (Experiment 1) Compared to exposure to a single dose of MA (5 mg/kg), chronic MA exposure decreased the number of c-Fos expressing cells in the paraventricular hypothalamus, dorsomedial hypothalamus, central amygdala, basolateral amygdala, bed nucleus of the stria terminalis (BNST), and CA3 hippocampal region. (Experiment 2) Compared to mice receiving their first dose of MA, mice chronically treated with MA, withdrawn, and re-administered MA, showed decreased c-Fos expressing cells within the central and basolateral amygdala, BNST, and CA3. CONCLUSIONS: HPA axis-associated amygdala, extended amygdala, and hippocampal regions endure lasting effects following chronic MA exposure and therefore may be linked to stress-related withdrawal symptoms.


Assuntos
Estimulantes do Sistema Nervoso Central/farmacologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Metanfetamina/farmacologia , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Tonsila do Cerebelo/citologia , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Animais , Temperatura Corporal/efeitos dos fármacos , Região CA3 Hipocampal/citologia , Região CA3 Hipocampal/efeitos dos fármacos , Região CA3 Hipocampal/metabolismo , Corticosterona/sangue , Sistema Hipotálamo-Hipofisário/citologia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/citologia , Proteínas Proto-Oncogênicas c-fos/biossíntese , Núcleos Septais/citologia , Núcleos Septais/efeitos dos fármacos , Núcleos Septais/metabolismo
16.
Behav Brain Res ; 298(Pt B): 1-11, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26522840

RESUMO

The brain might be exposed to irradiation under a variety of situations, including clinical treatments, nuclear accidents, dirty bomb scenarios, and military and space missions. Correctly recalling tasks learned prior to irradiation is important but little is known about post-learning effects of irradiation. It is not clear whether exposure to X-ray irradiation during memory consolidation, a few hours following training, is associated with altered contextual fear conditioning 24h after irradiation and which brain region(s) might be involved in these effects. Brain immunoreactivity patterns of the immediately early gene c-Fos, a marker of cellular activity was used to determine which brain areas might be altered in post-training irradiation memory retention tasks. In this study, we show that post-training gamma irradiation exposure (1 Gy) enhanced contextual fear memory 24h later and is associated with reduced cellular activation in the infralimbic cortex. Reduced GABA-ergic neurotransmission in parvalbumin-positive cells in the infralimbic cortex might play a role in this post-training radiation-enhanced contextual fear memory.


Assuntos
Córtex Cerebral/efeitos da radiação , Medo/efeitos da radiação , Raios gama/efeitos adversos , Memória/efeitos da radiação , Neurônios/efeitos da radiação , Animais , Córtex Cerebral/fisiopatologia , Condicionamento Psicológico/fisiologia , Condicionamento Psicológico/efeitos da radiação , Medo/fisiologia , Imuno-Histoquímica , Masculino , Memória/fisiologia , Camundongos Endogâmicos C57BL , Neurônios/fisiologia , Parvalbuminas/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Distribuição Aleatória , Ácido gama-Aminobutírico/metabolismo
17.
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
18.
J Neurosci ; 35(10): 4190-202, 2015 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-25762666

RESUMO

Pyramidal neurons in the medial prefrontal cortex (mPFC) critically contribute to cocaine-seeking behavior in humans and rodents. Activity of these neurons is significantly modulated by GABAergic, parvalbumin-containing, fast-spiking interneurons, the majority of which are enveloped by specialized structures of extracellular matrix called perineuronal nets (PNNs), which are integral to the maintenance of many types of plasticity. Using a conditioned place preference (CPP) procedure, we found that removal of PNNs primarily from the prelimbic region of the mPFC of adult, male, Sprague Dawley rats impaired the acquisition and reconsolidation of a cocaine-induced CPP memory. This impairment was accompanied by a decrease in the number of c-Fos-positive cells surrounded by PNNs. Following removal of PNNs, the frequency of inhibitory currents in mPFC pyramidal neurons was decreased; but following cocaine-induced CPP, both frequency and amplitude of inhibitory currents were decreased. Our findings suggest that cocaine-induced plasticity is impaired by removal of prelimbic mPFC PNNs and that PNNs may be a therapeutic target for disruption of cocaine CPP memories.


Assuntos
Lesões Encefálicas/complicações , Condicionamento Operante/fisiologia , Transtornos da Memória/etiologia , Rede Nervosa/fisiologia , Córtex Pré-Frontal/patologia , Animais , Aprendizagem por Associação/efeitos dos fármacos , Lesões Encefálicas/patologia , Condroitina ABC Liase/administração & dosagem , Cocaína/administração & dosagem , Condicionamento Operante/efeitos dos fármacos , Inibidores da Captação de Dopamina/administração & dosagem , Extinção Psicológica/efeitos dos fármacos , Extinção Psicológica/fisiologia , Masculino , Microscopia Confocal , Rede Nervosa/efeitos dos fármacos , Rede Nervosa/lesões , Proteínas do Tecido Nervoso/metabolismo , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Lectinas de Plantas/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de N-Acetilglucosamina/metabolismo , Fatores de Tempo
19.
J Neurochem ; 129(3): 495-508, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24400874

RESUMO

Dysregulation of hypothalamic-pituitary-adrenal (HPA) axis activation is associated with changes in addiction-related behaviors. In this study, we tested whether sex differences in the acute effects of methamphetamine (MA) exposure involve differential activation of the HPA axis. Male and female mice were injected with MA (1 mg/kg) or saline for comparison of plasma corticosterone and analysis of the immediate early gene c-Fos in brain. There was a prolonged elevation in corticosterone levels in female compared to male mice. C-Fos was elevated in both sexes following MA in HPA axis-associated regions, including the hypothalamic paraventricular nucleus (PVN), central amygdala, cingulate, and CA3 hippocampal region. MA increased the number of c-Fos and c-Fos/glucocorticoid receptor (GR) dual-labeled cells to a greater extent in males than females in the cingulate and CA3 regions. MA also increased the number of c-fos/vasopressin dual-labeled cells in the PVN as well as the number and percentage of c-Fos/GR dual-labeled cells in the PVN and central amygdala, although no sex differences in dual labeling were found in these regions. Thus, sex differences in MA-induced plasma corticosterone levels and activation of distinct brain regions and proteins involved in HPA axis regulation may contribute to sex differences in acute effects of MA on the brain. Methamphetamine induces a prolonged plasma corticosterone response in females compared to males. This may be mediated by increased neural activation, involving a greater activation of glucocorticoid receptor-positive cells, in males in the CA3 and cingulate brain regions, which are involved in negative feedback functions. These findings indicate a sex difference in the neural regulation of methamphetamine-induced plasma corticosterone release.


Assuntos
Estimulantes do Sistema Nervoso Central/farmacologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Metanfetamina/farmacologia , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Caracteres Sexuais , Animais , Corticosterona/sangue , Feminino , Sistema Hipotálamo-Hipofisário/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sistema Hipófise-Suprarrenal/metabolismo , Proteínas Proto-Oncogênicas c-fos/biossíntese , Radioimunoensaio
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA