Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Neuroscience ; 463: 184-196, 2021 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-33838289

RESUMO

Neuronal excitability and susceptibility to excitotoxic damage can be sex-specific, with neurons from males usually being more 'easily excitable' compared to neurons from females, especially during development. Increased excitability at an individual neuronal level can lead to the formation of hyperexcitable neuronal networks, which, consequently can make the brain more seizure prone. Both animal and clinical data suggest that males experience more frequent and severe seizures than do females. Serotonin (5-hydroxytryptamine; 5-HT) can mediate neuronal excitability and seizure behavior, often serving as an anticonvulsant. Importantly, 5-HT signaling during parts of the perinatal period is sexually dimorphic. Sex differences during development have been reported in both serotonin levels and receptor type (excitatory vs. inhibitory) expression in a manner that may leave the male brain more vulnerable to over-excitation. Thus, we aimed to determine if the anticonvulsant effects of 5-HT were sex- and/or age-dependent in juvenile animals. We report a baseline sex difference in N-methyl-d-aspartate (NMDA)-induced seizure behavior and hippocampal neuronal loss, with postnatal day (PND) 14 males exhibiting more severe seizure behavior compared to females. Pretreatment with the general 5-HT receptor agonist 5-methoxytryptamine (5-MT) abolishes baseline sex differences, providing an anticonvulsant effect for males only. These sex differences appear to be at least in part organized by testosterone, as females given neonatal androgen exhibit a seizure behavior profile in between that of males and females.


Assuntos
N-Metilaspartato/efeitos adversos , Convulsões/tratamento farmacológico , Serotonina/uso terapêutico , Animais , Feminino , Masculino , Gravidez , Ratos , Receptores de N-Metil-D-Aspartato , Convulsões/induzido quimicamente , Caracteres Sexuais
3.
Horm Behav ; 126: 104822, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32730760

RESUMO

Oxytocin is important for postnatal developmental experiences for mothers, infants, and transactions between them. Oxytocin is also implicated in adult affiliative behaviors, including social buffering of stress. There is evidence for connections between early life experience and adult oxytocin system functioning, but effects of early experience on behavioral, endocrine, and neurophysiological outcomes related to adult social buffering are not well explored. We use a limited bedding and nesting (LBN) material paradigm as an environmental disruption of early experiences and assessed central oxytocin systems in brain regions related to hypothalamic-pituitary-adrenal (HPA) axis regulation (paraventricular nucleus of the hypothalamus, amygdala, hippocampus). We also assessed developmentally-appropriate social behaviors and HPA reactivity during social buffering testing in adulthood. LBN litters had larger huddles and more pups visible compared to control litters during the first two weeks of life. LBN also altered the developmental trajectory of oxytocin-expressing cells and oxytocin receptor cells, with increases in oxytocin receptor cells at P15 in LBN pups. By adulthood, LBN females had more and LBN males had fewer oxytocin and oxytocin receptor cells in these areas compared to sex-matched controls. Adult LBN females, but not LBN males, had behavioral changes during social interaction and social buffering testing. The sex-specific effects of early experience on central oxytocin systems and social behavior may contribute to female resilience to early life adversity.


Assuntos
Sistema Hipotálamo-Hipofisário , Comportamento Materno/fisiologia , Ocitocina/metabolismo , Sistema Hipófise-Suprarrenal , Receptores de Ocitocina/metabolismo , Animais , Animais Recém-Nascidos , Comportamento Animal/fisiologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Feminino , Sistema Hipotálamo-Hipofisário/crescimento & desenvolvimento , Sistema Hipotálamo-Hipofisário/metabolismo , Masculino , Comportamento de Nidação/fisiologia , Ocitocina/farmacologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Sistema Hipófise-Suprarrenal/crescimento & desenvolvimento , Sistema Hipófise-Suprarrenal/metabolismo , Ratos , Ratos Long-Evans , Caracteres Sexuais , Comportamento Social , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia , Estresse Psicológico/prevenção & controle
4.
Brain Behav ; 9(5): e01265, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30912298

RESUMO

INTRODUCTION: Perineuronal nets (PNNs) are extracellular matrices that encompass parvalbumin-expressing parvalbumin positive (PVALB+) fast-spiking inhibitory interneurons where they protect and stabilize afferent synapses. Recent observations that gonadal hormones influence PVALB+ neuron development suggest that PNN regulation may be sexually dimorphic. Sex differences in PNN abundance and complexity have been reported in sexually dimorphic nuclei in zebra finch brains; however, corresponding differences in mammalian brains have not been investigated. METHODS: In this study we assessed the number of cortical and hippocampal PNNs in juvenile and young adult male and female rats using fluorescent immunohistochemistry for PVALB and the PNN marker Wisteria Floribunda Lectin. RESULTS: We report here that PNNs are numerous and well developed in hippocampal cornu ammonis-1 of adult males but are lower in juvenile and possibly adult females. No significant differences were observed between sexes in cornu ammonis-3 or adjacent neocortex. There was an observed developmental difference in the neocortex as juveniles had more PVALB+ cells, but fewer PNN+ cells, than adults. CONCLUSIONS: Because PNNs are integral for several hippocampal-mediated learning and memory tasks, these observations have potential sex-dependent translational implications for clinical strategies targeting cognitive dysfunction.


Assuntos
Interneurônios/fisiologia , Parvalbuminas/metabolismo , Caracteres Sexuais , Fatores Etários , Animais , Comportamento Animal/fisiologia , Região CA1 Hipocampal/metabolismo , Matriz Extracelular/metabolismo , Feminino , Imuno-Histoquímica , Masculino , Ratos , Lobo Temporal/metabolismo
5.
Brain Res ; 1701: 85-92, 2018 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-30040918

RESUMO

Perinatal administration of serotonin (5HT) agonist 5-methoxytryptamine (5MT) induces developmental hyperserotonemia (DHS; elevated blood serotonin) and produces behavioral and neurochemical changes in rats relevant to Autism Spectrum Disorder (ASD), such as oxytocin dysregulation. Disruption of the oxytocin system may underlie many of the social deficits present in ASD individuals, thus we investigated the mechanism(s) underlying DHS-induced oxytocin dysregulation. The most parsimonious mechanism of 5HT action would be alteration of 5HT receptors on oxytocin cells; 5HT is known to influence cell survival as well as influence oxytocin release via 5HT1A and 5HT2A receptors, which co-localize in oxytocin-expressing (OXT+) cells in the paraventricular nucleus (PVN) of the hypothalamus. We report that both male and female DHS rats have a lower percentage of OXT+ cells co-localized with excitatory 5HT2A receptors than control animals, while only DHS females have a higher percentage of OXT+ cells co-localized with inhibitory 5HT1A receptors compared to controls. Importantly, DHS also reduces the number of OXT+ cells in the PVN of adult male, but not female, rats. This pattern suggests that females, but not males, can regulate 5HT receptors in response to DHS in a manner that promotes oxytocin cell survival and functional efficiency. In addition, it has been previously reported that DHS alters normal juvenile play, especially in males, thus we also tested play partner preference among juvenile control and DHS males. Sex differences observed using the DHS model of ASD add to its validity, given the pronounced male sex bias in the prevalence of ASD, and emphasize the need for inclusion of both sexes in ASD research.


Assuntos
Ocitocina/metabolismo , Receptor 5-HT1A de Serotonina/metabolismo , Receptores de Serotonina/metabolismo , 5-Metoxitriptamina/farmacologia , Animais , Transtorno do Espectro Autista/sangue , Transtorno Autístico/sangue , Transtorno Autístico/metabolismo , Modelos Animais de Doenças , Feminino , Hipotálamo/efeitos dos fármacos , Masculino , Ocitocina/sangue , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Serotonina/fisiologia , Serotonina/metabolismo , Caracteres Sexuais , Fatores Sexuais , Comportamento Sexual
6.
Front Neuroendocrinol ; 42: 40-52, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26930421

RESUMO

Hormones have wide-ranging effects throughout the nervous system, including the ability interact with and modulate many aspects of intracellular calcium regulation and calcium signaling. Indeed, these interactions specifically may help to explain the often opposing or paradoxical effects of hormones, such as their ability to both promote and prevent neuronal cell death during development, as well as reduce or exacerbate damage following an insult or injury in adulthood. Here, we review the basic mechanisms underlying intracellular calcium regulation-perhaps the most dynamic and flexible of all signaling molecules-and discuss how gonadal hormones might manipulate these mechanisms to coordinate diverse cellular responses and achieve disparate outcomes. Additional future research that specifically addresses questions of sex and hormone effects on calcium signaling at different ages will be critical to understanding hormone-mediated neuroprotection.


Assuntos
Sinalização do Cálcio/fisiologia , Hormônios Gonadais/fisiologia , Neuroproteção/fisiologia , Animais , Humanos
7.
Dev Neurobiol ; 76(11): 1241-1253, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-26899026

RESUMO

Testosterone and its metabolites masculinize the brain during a critical perinatal window, including the relative volume of sexually dimorphic brain areas such as the sexually dimorphic nucleus of the preoptic area (SDN), which is larger in males than females. Serotonin (5HT) may mediate this hormone action, since 5HT given during the second week of life decreases (i.e., feminizes) SDN volume in males and testosterone-treated females. Although previous work indicates that the 5HT2A/2C receptor is sufficient to induce feminization, it is unclear whether other serotonin receptors are required and which subpopulation(s) of SDN cells are specifically organized by 5HT. Therefore, we injected male and female Sprague-Dawley rat pups with saline, a nonselective 5HTR agonist, a 5HT2A/2C agonist, or a 5HT2A/2C antagonist over several timecourses in early life, and measured the Nissl-SDN as well as a calbindin+ subdivision of the SDN, the CALB-SDN. When examined on postnatal day 18 or early adulthood, the size of the Nissl-SDN was feminized in males treated with any of the serotonergic drugs, eliminating the typical sex difference. In contrast, the sex difference in CALB-SDN size was maintained regardless of serotoninergic drug treatment. This pattern suggests that although gonadal hormones shape the whole SDN, individual cellular phenotypes respond to different intermediary signals to become sexually dimorphic. Specifically, 5HT mediates sexual differentiation of non-calbindin population(s) within the SDN. The results also caution against using measurement of the CALB-SDN in isolation, as the absence of an effect on the CALB-SDN does not preclude an effect on the overall nucleus. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 76: 1241-1253, 2016.


Assuntos
Calbindinas/metabolismo , Feminização/metabolismo , Área Pré-Óptica/metabolismo , Serotonina/fisiologia , Diferenciação Sexual/fisiologia , Animais , Calbindinas/efeitos dos fármacos , Feminino , Masculino , Gravidez , Área Pré-Óptica/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Serotonina/metabolismo , Antagonistas da Serotonina/farmacologia , Agonistas do Receptor de Serotonina/farmacologia , Diferenciação Sexual/efeitos dos fármacos
8.
Physiol Behav ; 128: 260-9, 2014 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-24530263

RESUMO

There is a striking sex difference in the diagnosis of Autism Spectrum Disorder (ASD), such that males are diagnosed more often than females, usually in early childhood. Given that recent research has implicated elevated blood serotonin (hyperserotonemia) in perinatal development as a potential factor in the pathogenesis of ASD, we sought to evaluate the effects of developmental hyperserotonemia on social behavior and relevant brain morphology in juvenile males and females. Administration of 5-methoxytryptamine (5-MT) both pre- and postnatally was found to disrupt normal social play behavior in juveniles. In addition, alterations in the number of oxytocinergic cells in the lateral and medial paraventricular nucleus (PVN) were evident on postnatal day 18 (PND18) in 5-MT treated females, but not treated males. 5-MT treatment also changed the relative expression of 5-HT(1A) and 5-HT(2A) receptors in the PVN, in males at PND10 and in females at PND18. These data suggest that serotonin plays an organizing role in the development of the PVN in a sexually dimorphic fashion, and that elevated serotonin levels during perinatal development may disrupt normal organization, leading to neurochemical and behavioral changes. Importantly, these data also suggest that the inclusion of both juvenile males and females in studies will be necessary to fully understand the role of serotonin in development, especially in relation to ASD.


Assuntos
Hipotálamo/fisiologia , Receptores de Ocitocina/fisiologia , Receptores de Serotonina/fisiologia , Fatores Etários , Animais , Comportamento Animal/fisiologia , Feminino , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Masculino , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/fisiologia , Jogos e Brinquedos , Ratos , Ratos Sprague-Dawley/crescimento & desenvolvimento , Receptor 5-HT2A de Serotonina/biossíntese , Receptor 5-HT2A de Serotonina/fisiologia , Receptores de Ocitocina/biossíntese , Receptores de Serotonina/biossíntese , Serotonina/sangue , Serotonina/farmacologia , Fatores Sexuais
9.
PLoS One ; 8(7): e69672, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23874981

RESUMO

Male sexual behavior (MSB) is modulated by gonadal steroids, yet this relationship is highly variable across species and between individuals. A significant percentage (~30%) of B6D2F1 hybrid male mice demonstrate MSB after long-term orchidectomy (herein after referred to as "maters"), providing an opportunity to examine the mechanisms that underlie individual differences in steroidal regulation of MSB. Use of gene expression arrays comparing maters and non-maters has provided a first pass look at the genetic underpinnings of steroid-independent MSB. Surprisingly, of the ~500 genes in the medial preoptic area (MPOA) that differed between maters and non-maters, no steroid hormone or receptor genes were differentially expressed between the two groups. Interestingly, best known for their association with Alzheimer's disease, amyloid precursor protein (APP) and the microtubule-associated protein tau (MAPT) were elevated in maters. Increased levels of their protein products (APP and tau) in their non-pathological states have been implicated in cell survival, neuroprotection, and supporting synaptic integrity. Here we tested transgenic mice that overexpress tau and found facilitated mounting and intromission behavior after long-term orchidectomy relative to littermate controls. In addition, levels of synaptophysin and spinophilin, proteins generally enriched in synapses and dendritic spines respectively, were elevated in the MPOA of maters. Dendritic morphology was also assessed in Golgi-impregnated brains of orchidectomized B6D2F1 males, and hybrid maters exhibited greater dendritic spine density in MPOA neurons. In sum, we show for the first time that retention of MSB in the absence of steroids is correlated with morphological differences in neurons.


Assuntos
Espinhas Dendríticas/metabolismo , Comportamento Sexual Animal/fisiologia , Esteroides/metabolismo , Proteínas tau/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Feminino , Individualidade , Masculino , Camundongos
10.
Psychoneuroendocrinology ; 35(2): 197-208, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19589643

RESUMO

Methamphetamine (MA) abuse has reached epidemic proportions in the United States. Users of MA report dramatic increases in sexual drive that have been associated with increased engagement in risky sexual behavior leading to higher rates of sexually transmitted diseases and unplanned pregnancies. The ability of MA to enhance sexual drive in females is enigmatic since related psychostimulants like amphetamine and cocaine appear not to affect sexual drive in women, and in rodents models, amphetamine has been reported to be inhibitory to female sexual behavior. Examination of MA's effects on female sexual behavior in an animal model is lacking. Here, using a rodent model, we have demonstrated that MA enhanced female sexual behavior. MA (5mg/kg) or saline vehicle was administered once daily for 3 days to adult ovariectomized rats primed with ovarian steroids. MA treatment significantly increased the number of proceptive events and the lordosis response compared to hormonally primed, saline controls. The effect of MA on the neural circuitry underlying the motivation for sexual behavior was examined using Fos immunoreactivity. In the medial amygdala and the ventromedial nucleus of the hypothalamus, nuclei implicated in motivated behaviors, ovarian hormones and MA independently enhance the neuronal activation, but more striking was the significantly greater activation induced by their combined administration. Increases in dopamine neurotransmission may underlie the MA/hormone mediated increase in neuronal activation. In support of this possibility, ovarian hormones significantly increased tyrosine hydroxylase (the rate limiting enzyme in dopamine synthesis) immunoreactivity in the medial amygdala. Thus our present data suggest that the interactions of MA and ovarian hormones leads to changes in the neural substrate of key nuclei involved in mediating female sexual behaviors, and these changes may underlie MA's ability to enhance these behaviors.


Assuntos
Tonsila do Cerebelo/efeitos dos fármacos , Metanfetamina/farmacologia , Neurônios/efeitos dos fármacos , Comportamento Sexual Animal/efeitos dos fármacos , Núcleo Hipotalâmico Ventromedial/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Tonsila do Cerebelo/fisiologia , Animais , Estimulantes do Sistema Nervoso Central/farmacologia , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Feminino , Hipercinese/induzido quimicamente , Neurônios/fisiologia , Proteínas Oncogênicas v-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Caracteres Sexuais , Transmissão Sináptica/efeitos dos fármacos , Tirosina 3-Mono-Oxigenase/metabolismo , Núcleo Hipotalâmico Ventromedial/metabolismo , Núcleo Hipotalâmico Ventromedial/fisiologia
11.
Physiol Behav ; 96(2): 383-8, 2009 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-18976678

RESUMO

Processing of relevant olfactory and pheromonal cues has long been known as an important process necessary for social and sexual behavior in rodents. Several nuclei that receive input from the vomeronasal projection pathway are involved in sexual behavior and show changes in immediate early gene expression after stimulation with a variety of sex-related stimuli. The nuclei in this pathway are sexually dimorphic due to the early patterning events induced by estradiol derived from testicular androgens, which developmentally defeminize and masculinize the brain and adult sexual behavior. Masculinization can be induced independently of estradiol via prostaglandin-E(2) (PGE(2)), and therefore assessed separately from defeminization. Here we examined the effects of brain defeminization and masculinization on neuronal response to sex-related odors using Fos, the protein product of the immediate early gene c-fos, as an indicator of activity. Female rat pups treated with a cyclooxygenase-2 inhibitor, to reduce PGE(2), plus estradiol, estradiol alone, and PGE(2) alone were exposed to estrous female odor as adults and the resulting Fos expression was examined in the medial amygdala, preoptic area, and ventromedial nucleus of the hypothalamus. Defeminized and/or masculinized females all showed patterns of Fos activity similar to control males and significantly different from control females. These results suggest that early exposure to estradiol and PGE(2) do not affect olfaction in females, but switch the activity pattern of sex-related nuclei in females to resemble that of males following exposure to sexually-relevant cues.


Assuntos
Encéfalo/efeitos dos fármacos , Dinoprostona/farmacologia , Estradiol/farmacologia , Estrogênios/farmacologia , Ciclo Estral , Odorantes , Proteínas Proto-Oncogênicas c-fos/metabolismo , Fatores Etários , Análise de Variância , Animais , Animais Recém-Nascidos , Encéfalo/metabolismo , Dinoprostona/metabolismo , Feminino , Masculino , Ovariectomia/métodos , Gravidez , Ratos , Ratos Sprague-Dawley , Estatísticas não Paramétricas
12.
J Neurosurg Anesthesiol ; 20(4): 233-40, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18812886

RESUMO

Certain anesthetics exhibit neurotoxicity in the brains of immature but not mature animals. Gamma-aminobutyric acid (GABA), the primary inhibitory neurotransmitter in the adult brain, is excitatory on immature neurons via its action at the GABAA receptor, due to a reversed transmembrane chloride gradient. GABAA receptor activation in immature neurons is sufficient to open L-type voltage-gated calcium channels. As propofol is a GABAA agonist, we hypothesized that it and more specific GABAA modulators would increase intracellular free calcium ([Ca2+]i), resulting in the death of neonatal rat hippocampal neurons. Neuronal [Ca2+]i was monitored using Fura2-AM fluorescence imaging. Cell death was assessed by double staining with propidium iodide and Hoechst 33258 at 1 hour (acute) and 48 hours (delayed) after 5 hours exposure of neurons to propofol or the GABAA receptor agonist, muscimol, in the presence and absence of the GABA receptor antagonist, bicuculline, or the L-type Ca2+ channel blocker, nifedipine. Fluorescent measurements of caspase-3,-7 activities were performed at 1 hour after exposure. Both muscimol and propofol induced a rapid increase in [Ca2+]i in days in vitro (DIV) 4, but not in DIV 8 neurons, that was inhibited by nifedipine and bicuculline. Caspase-3,-7 activities and cell death increased significantly in DIV 4 but not DIV 8 hippocampal neuronal cultures 1 hour after 5 hours exposure to propofol, but not muscimol, and were inhibited by the presence of bicuculline or nifedipine. We conclude that an increase in [Ca2+]i, due to activation of GABAA receptors and opening of L-type calcium channels, is necessary for propofol-induced death of immature rat hippocampal neurons but that additional mechanisms not elicited by GABAA activation alone also contribute to cell death.


Assuntos
Anestésicos Intravenosos/toxicidade , Neurônios/efeitos dos fármacos , Propofol/toxicidade , Ácido gama-Aminobutírico/fisiologia , Envelhecimento/fisiologia , Animais , Cálcio/metabolismo , Agonistas dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 7/metabolismo , Morte Celular/efeitos dos fármacos , Células Cultivadas , Eletrofisiologia , Agonistas GABAérgicos/farmacologia , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Microscopia de Fluorescência , Muscimol/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de GABA-A/efeitos dos fármacos
13.
Eur J Neurosci ; 27(4): 791-800, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18333959

RESUMO

The hippocampus is a key brain region regulating complex cognitive and emotional responses, and is implicated in the etiology of depressive and anxiety disorders, many of which exhibit some degree of sex difference. The male rat hippocampus is consistently reported to be slightly but significantly larger than the female. The majority of studies on the development of volumetric sex differences have focused on the effects of estradiol (E2), with relatively few focusing on androgens. We examined the impact of both E2 and androgens on newly born cells in the developing rat hippocampus, and report that neonatal males have significantly more 5-bromo-2'-deoxyuridine-5'-monophosphate (BrdU)+ cells than females. Both testosterone (T) and dihydrotestosterone treatment of females significantly increased the number of BrdU+ cells, an effect blocked by the androgen receptor antagonist, flutamide. However, only T significantly increased the number of neuronal nuclear antigen+ neurons in the female rat hippocampus. Interestingly, E2 treatment also increased BrdU+ cells in females, but had no effect on neuron number. Instead, E2 and T significantly increased the number of newly born glial fibrillary acidic protein or glutamine synthetase+ glial cells in females, indicating that androgens and E2 may act independently to achieve distinct endpoints. Quantification of pyknotic cells at two different developmental time points indicates no sex difference in the number of cells dying, suggesting, but not proving, that gonadal steroids are promoting cell genesis.


Assuntos
Androgênios/farmacologia , Estradiol/farmacologia , Hipocampo/crescimento & desenvolvimento , Neuroglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Caracteres Sexuais , Animais , Contagem de Células , Proliferação de Células , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Imuno-Histoquímica , Masculino , Neuroglia/citologia , Neuroglia/fisiologia , Neurônios/citologia , Neurônios/fisiologia , Ratos , Ratos Sprague-Dawley
14.
Endocrinology ; 148(7): 3391-401, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17412802

RESUMO

Steroid-mediated sexual differentiation of the brain is a developmental process that permanently organizes the brain into a male or female phenotype. Previous studies in the rodent have examined the steroid-mediated mechanisms of male brain development. In an effort to identify molecules involved in female brain development, a high-throughput proteomics approach called PowerBlot was used to identify signaling proteins differentially regulated in the neonatal male and female rat hypothalamus during the critical period for brain sexual differentiation. Focal adhesion kinase (FAK) and paxillin, both members of the focal adhesion complex family of proteins, were significantly elevated in the newborn female compared with the male hypothalamus. Sex differences in these proteins were not detected in brain regions that are not subject to substantial organizational effects of steroids. Estrogens, the aromatized products of testosterone in the male, can both masculinize and defeminize the male brain. Daily estradiol administration to neonatal females significantly reduced FAK and paxillin in the hypothalamus, and aromatase inhibition increased paxillin in males to levels comparable with females. Androgens also appear to modulate paxillin levels in combination with estrogen action. Across development, hypothalamic levels of FAK were significantly elevated in females compared with males on postnatal d 6. Synaptic circuits in the hypothalamus develop sex differences perinatally. Estradiol treatment of cultured hypothalamic neurons significantly enhanced axon branching (P<0.01), consistent with the phenotype of FAK-deficient neurons. Together, these data implicate FAK and paxillin as regulators of sex differences in neuronal morphology.


Assuntos
Encéfalo/enzimologia , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Paxilina/metabolismo , Diferenciação Sexual , Animais , Animais Recém-Nascidos , Aromatase/efeitos dos fármacos , Western Blotting , Encéfalo/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Células Cultivadas , Estradiol/farmacologia , Feminino , Hipotálamo/citologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/enzimologia , Imuno-Histoquímica , Letrozol , Masculino , Neuritos/efeitos dos fármacos , Nitrilas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Estrogênio/antagonistas & inibidores , Fatores Sexuais , Tamoxifeno/farmacologia , Testosterona/farmacologia , Triazóis/farmacologia
15.
Endocrine ; 29(2): 217-31, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16785598

RESUMO

Ovarian hormones can protect against brain injury, neurodegeneration, and cognitive decline. Most attention has focused on estrogens and accumulating data demonstrate that estrogen seems to specifically protect cortical and hippocampal neurons from ischemic injury and from damage due to severe seizures. Although multiple studies demonstrate protection by estrogen, in only a few instances is the issue of how the steroid confers protection known. Here, we first review data evaluating the neuroprotective effects of estrogens, a selective estrogen receptor modulator (SERM), and estrogen receptor alpha- and beta-selective ligands in animal models of focal and global ischemia. Using focal ischemia in ovariectomized ERalphaKO, ERbetaKO, and wild-type mice, we clearly established that the ERalpha subtype is the critical ER mediating neuroprotection in mouse focal ischemia. In rats and mice, the middle cerebral artery occlusion (MCAO) model was used to represent cerebrovascular stroke, while in gerbils the two-vessel occlusion model, representing global ischemia, was used. The gerbil global ischemia model was used to evaluate the neuroprotective effects of estrogen, SERMs, and ERalpha- and ERbeta-selective compounds in the hippocampus. Analysis of neurogranin mRNA, a marker of viability of hippocampal neurons, with in situ hybridization, revealed that estrogen treatment protected the dorsal CA1 regions not only when administered before, but also when given 1 h after occlusion. Estrogen rarely is secreted alone and studies of neuroprotection have been less extensive for a second key ovarian hormone progesterone. In the second half of this review, we present data on neuroprotection by estrogen and progesterone in animal model of epilepsy followed by exploration into ovarian steroid effects on neuronal damage in models of multiple sclerosis and traumatic brain injury.


Assuntos
Estrogênios/farmacologia , Doenças do Sistema Nervoso/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Ovário/metabolismo , Progesterona/farmacologia , Animais , Lesões Encefálicas/prevenção & controle , Isquemia Encefálica/prevenção & controle , Citocinas/efeitos adversos , Epilepsia , Feminino , Modelos Animais , Modelos Biológicos , Esclerose Múltipla/prevenção & controle , Receptores de Esteroides/metabolismo
17.
J Neurosci ; 23(6): 2357-62, 2003 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-12657695

RESUMO

Several sex differences in the nervous system depend on differential cell death during development in males and females. The anti-apoptotic protein, Bcl-2, promotes the survival of many types of neurons during development and in response to injury. To determine whether Bcl-2 might similarly control cell death in sexually dimorphic regions, we compared neuron number in wild-type mice and transgenic mice overexpressing Bcl-2 under the control of a neuron-specific promoter. Three neural areas were examined: the spinal nucleus of the bulbocavernosus (SNB), in which neuron number is greater in males; the retrodorsolateral nucleus (RDLN) of the spinal cord, which exhibits no sex difference in neuron number; and the anteroventral periventricular nucleus (AVPV) of the hypothalamus, in which both overall cell density and the number of tyrosine hydroxylase immunoreactive (TH-ir) neurons are greater in females. Bcl-2 overexpression significantly increased SNB cell number in females, overall cell density of AVPV in males, and RDLN cell number in both sexes. Bcl-2 overexpression did not alter the number of TH-ir neurons in AVPV of males or females. These findings indicate that Bcl-2 can regulate sexually dimorphic cell number in the brain and spinal cord and suggest that Bcl-2 may mediate effects of testosterone on cell survival during neural development. In contrast to the regulation of overall cell density in AVPV, the sex difference in TH cell number apparently is not caused by a Bcl-2-dependent mechanism.


Assuntos
Encéfalo/citologia , Neurônios/citologia , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Caracteres Sexuais , Medula Espinal/citologia , Animais , Contagem de Células , Tamanho Celular , Feminino , Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Neurônios Motores/citologia , Neurônios Motores/metabolismo , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Transgenes , Tirosina 3-Mono-Oxigenase/biossíntese
18.
Brain Res ; 950(1-2): 312-6, 2002 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-12231259

RESUMO

Bcl-2 and Bax immunoreactivity were examined in the spinal nucleus of the bulbocavernosus (SNB), an androgen-sensitive motor pool of adult rats. Castration reduced Bcl-2 immunoreactivity and testosterone treatment of castrates prevented this decline. Hormone manipulations did not affect Bcl-2 or Bax staining in the retrodorsolateral nucleus (RDLN), a relatively androgen-insensitive nucleus at the same spinal level. Changes in Bcl-2 expression may underlie the hormonal control of cell death and/or neural plasticity in SNB motoneurons.


Assuntos
Neurônios Motores/química , Proteínas Proto-Oncogênicas c-bcl-2/análise , Caracteres Sexuais , Testosterona/metabolismo , Envelhecimento/fisiologia , Animais , Feminino , Imuno-Histoquímica , Masculino , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/metabolismo , Orquiectomia/estatística & dados numéricos , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Ratos , Ratos Sprague-Dawley , Testosterona/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...