Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 136
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(43): e2306475120, 2023 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-37847733

RESUMO

Anxiety disorders are a major public health concern and current treatments are inadequate for many individuals. Anxiety is more common in women than men and this difference arises during puberty. Sex differences in physiological stress responses may contribute to this variability. During puberty, gonadal hormones shape brain structure and function, but the extent to which these changes affect stress sensitivity is unknown. We examined how pubertal androgens shape behavioral and neural responses to social stress in California mice (Peromyscus californicus), a model species for studying sex differences in stress responses. In adults, social defeat reduces social approach and increases social vigilance in females but not males. We show this sex difference is absent in juveniles, and that prepubertal castration sensitizes adult males to social defeat. Adult gonadectomy does not alter behavioral responses to defeat, indicating that gonadal hormones act during puberty to program behavioral responses to stress in adulthood. Calcium imaging in the medioventral bed nucleus of the stria terminalis (BNST) showed that social threats increased neural activity and that prepubertal castration generalized these responses to less threatening social contexts. These results support recent hypotheses that the BNST responds to immediate threats. Prepubertal treatment with the nonaromatizable androgen dihydrotestosterone acts in males and females to reduce the effects of defeat on social approach and vigilance in adults. These data indicate that activation of androgen receptors during puberty is critical for programming behavioral responses to stress in adulthood.


Assuntos
Núcleos Septais , Diferenciação Sexual , Adulto , Humanos , Masculino , Feminino , Androgênios/farmacologia , Hormônios Gonadais/farmacologia , Hormônios Gonadais/fisiologia , Puberdade
2.
Gene ; 782: 145529, 2021 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-33631246

RESUMO

Male sex differentiation in the crustacean is best known to be controlled by the insulin-like androgenic gland hormone (IAG). In this report, the cDNA and gene of the shrimp Fenneropenaeus merguiensis FmIAG were studied and characterized. FmIAG gene shares a high sequence identity in the coding region as well as the promoter region with that of F. chinensis. FmIAG gene is most likely consists of 5 exons and 4 introns. The cDNA reported here is the most abundant transcript that retained cryptic intron 4. The use of different splicing acceptor sites in exon 2 can produce a long-form FmIAG transcript variant with 6 additional amino acids inserted. Splicing of cryptic intron 4 would produce a transcript variant with a different C-terminal end. Therefore 4 different FmIAG transcripts can be produced from the FmIAG gene. During the molt cycle, the expression level of FmIAG was low in the early intermolt, increase steadily towards the late premolt and decreased rapidly in the early postmolt. In addition to the androgenic gland, FmIAG is also expressed in the hepatopancreas and ovary of adult females. Unilateral eyestalk ablation caused a significant increase in FmIAG transcript suggesting that the eyestalk consists of inhibiting factor(s) that suppressesFmIAGexpression. To explore the function of FmIAG in males, injection of FmIAG dsRNA knock-down the expression of FmIAG and up-regulated the expression of the vitellogenin gene in the testis and hepatopancreas. Interestingly a CHH-like gene identified in the androgenic gland was down-regulated. CHH-like gene knock-down resulted in altered expression of FmIAG in males suggesting that the CHH-like may be involved in FmIAG's regulation. RT-PCR with specific primers to the different transcript variant were used to determine if there is an association of different sizes of male and the type of IAG transcript. Results indicated that a high percentage of the large male shrimp expressed the long-form of FmIAG. The results suggested that FmIAG may be useful as a size marker for male shrimp aquaculture. In summary, the results of this study have expanded our knowledge of shrimp insulin-like androgenic gland hormone in male sex development and its potential role as a marker gene for growth regulation in shrimp.


Assuntos
Hormônios Gonadais/genética , Hormônios de Invertebrado/genética , Penaeidae/genética , Processamento Alternativo , Animais , Proteínas de Artrópodes/genética , Proteínas de Artrópodes/fisiologia , DNA Complementar , Éxons , Feminino , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Variação Genética , Hormônios Gonadais/fisiologia , Hepatopâncreas/metabolismo , Íntrons , Hormônios de Invertebrado/fisiologia , Masculino , Muda/genética , Penaeidae/fisiologia , Filogenia , Diferenciação Sexual/genética
3.
Endocrinology ; 161(9)2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32735650

RESUMO

Ovarian hormones, including 17ß-estradiol, are implicated in numerous physiological processes, including sleep. Beginning at puberty, girls report more sleep complaints than boys, which is maintained throughout the reproductive life stage. Sleep problems are exacerbated during the menopausal transition, evidenced by greater risk for sleep disorders. There is emerging evidence that menopause-associated hormone loss contributes to this elevated risk, but age is also an important factor. The extent to which menopause-associated sleep disturbance persists into postmenopause above and beyond the effects of age remains unknown. Untreated sleep disturbances have important implications for cognitive health, as they are emerging as risk factors for dementia. Given that sleep loss impairs memory, an important knowledge gap concerns the role played by menopause-associated hormone loss in exacerbating sleep disturbance and, ultimately, cognitive function in aging women. In this review, we take a translational approach to illustrate the contribution of ovarian hormones in maintaining the sleep-wake cycle in younger and middle-aged females, with evidence implicating 17ß-estradiol in supporting the memory-promoting effects of sleep. Sleep physiology is briefly reviewed before turning to behavioral and neural evidence from young females linking 17ß-estradiol to sleep-wake cycle maintenance. Implications of menopause-associated 17ß-estradiol loss is also reviewed before discussing how ovarian hormones may support the memory-promoting effects of sleep, and why menopause may exacerbate pathological aging via effects on sleep. While still in its infancy, this research area offers a new sex-based perspective on aging research, with a focus on a modifiable risk factor for pathological aging.


Assuntos
Envelhecimento/efeitos dos fármacos , Hormônios Gonadais/farmacologia , Sono/efeitos dos fármacos , Adulto , Envelhecimento/fisiologia , Cognição/efeitos dos fármacos , Cognição/fisiologia , Feminino , Hormônios Gonadais/metabolismo , Hormônios Gonadais/fisiologia , Humanos , Menopausa/efeitos dos fármacos , Menopausa/fisiologia , Ciclo Menstrual/efeitos dos fármacos , Ciclo Menstrual/fisiologia , Ovário/metabolismo , Pós-Menopausa/efeitos dos fármacos , Pós-Menopausa/fisiologia , Sono/fisiologia
4.
Front Neuroendocrinol ; 58: 100853, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32640267

RESUMO

It is well known that stroke incidence and outcome is sex-dependent and influenced by age and gonadal hormones. In post-menopausal and/or aged females, declining estrogen levels increases stroke risk. However, women who experience early menopause also have an increase in stroke risk. This suggests that, regardless of age, gonadal hormones regulate stroke risk and severity. This review discusses prolonged gonadal hormone dysfunction in a common female endocrine disorder known as polycystic ovarian syndrome, PCOS, and the associated increased risk of stroke due to resulting hyperandrogenism and metabolic comorbidities.


Assuntos
Hormônios Gonadais/fisiologia , Síndrome do Ovário Policístico/complicações , Acidente Vascular Cerebral/etiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/fisiologia , Feminino , Hormônios Gonadais/sangue , Humanos , Hiperandrogenismo/sangue , Hiperandrogenismo/complicações , Hiperandrogenismo/epidemiologia , Pessoa de Meia-Idade , Síndrome do Ovário Policístico/sangue , Síndrome do Ovário Policístico/epidemiologia , Fatores de Risco , Índice de Gravidade de Doença , Acidente Vascular Cerebral/sangue , Acidente Vascular Cerebral/epidemiologia , Acidente Vascular Cerebral/patologia
5.
Neuroreport ; 31(10): 717-723, 2020 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-32453019

RESUMO

The brain's default mode network (DMN) has become closely associated with self-referential mental activity, particularly in the resting-state. Prior reports point that the sex hormones are potent modulators of brain plasticity and functional connectivity. However, it is uncertain whether changes in ovarian hormones, as occur during the monthly menstrual cycle, substantially affects the functional connectivity of DMN. Here, we employed a Self-Awareness Scale (SAS) and the resting-state functional MRI in the late follicular phase and the mid-luteal phase to investigate the effect of the estradiol (E2) and progesterone on the SAS and DMN. On the behavioral level, increased progesterone facilitated women's other-focused attention. The regions of interest-based resting-state functional connectivity analyses continued to demonstrate a negative correlation of the relative progesterone and the medial prefrontal cortex-inferior temporal gyrus (mPFC-ITG) functional connectivity, and a facilitated effect of relative E2 on the mPFC-inferior parietal lobule functional connectivity in the DMN. Furthermore, as a core hub of the 'theory of mind', the functional connectivity between the ITG and thalamus was found negatively correlated with the relative E2. Meanwhile, the mid-luteal phase, which had significantly lower relative E2 levels, was indicated had stronger ITG-thalamus functional connectivity during the resting state. These results demonstrated an opposite effect of E2 and progesterone on the DMN and the other-focused preference in the mid-luteal phase, extended previous evidence of the potentially adaptive psychological effects of ovarian hormones on mapping self and others in the brain networks.


Assuntos
Rede de Modo Padrão/fisiologia , Estrogênios/fisiologia , Ovário/fisiologia , Progesterona/fisiologia , Autoimagem , Adulto , Mapeamento Encefálico , Feminino , Fase Folicular/fisiologia , Hormônios Gonadais/fisiologia , Humanos , Fase Luteal/fisiologia , Imageamento por Ressonância Magnética , Adulto Jovem
6.
Psychoneuroendocrinology ; 109: 104387, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31465941

RESUMO

Sex differences in the brain and behavior are produced by the perinatal action of testosterone, which is converted into estradiol by the enzyme aromatase in the brain. Although magnetic resonance imaging (MRI) has been widely used in humans to study these differences, the use of animal models, where hormonal status can be properly manipulated, is necessary to explore the mechanisms involved. We used sheep, a recognized model in the field of neuroendocrinology, to assess brain morphological and functional sex differences and their regulation by adult gonadal hormones. To this end, we performed voxel-based morphometry and a resting-state functional MRI approach to assess sex differences in gonadally intact animals. We demonstrated significant sex differences in gray matter concentration (GMC) at the level of the gonadotropic axis, i.e., not only within the hypothalamus and pituitary but also within the hippocampus and the amygdala of intact animals. We then performed the same analysis one month after gonadectomy and found that some of these differences were reduced, especially in the hypothalamus and amygdala. By contrast, we found few differences in the organization of the functional connectome between males and females either before or after gonadectomy. As a whole, our study identifies brain regions that are sexually dimorphic in the sheep brain at the resolution of the MRI and highlights the role of gonadal hormones in the maintenance of these differences.


Assuntos
Encéfalo/efeitos dos fármacos , Hormônios Gonadais/metabolismo , Caracteres Sexuais , Tonsila do Cerebelo/metabolismo , Animais , Encéfalo/diagnóstico por imagem , Castração/métodos , Estradiol/farmacologia , Feminino , Hormônios Gonadais/fisiologia , Substância Cinzenta/diagnóstico por imagem , Substância Cinzenta/efeitos dos fármacos , Hipotálamo/metabolismo , Imageamento por Ressonância Magnética/métodos , Masculino , Ovinos , Testosterona/farmacologia
7.
Curr Opin Psychiatry ; 32(6): 484-490, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31306250

RESUMO

PURPOSE OF REVIEW: Females experience eating disorders at substantially higher rates than males. Although sociocultural factors have traditionally been thought to underlie this sex disparity, accumulating evidence implicates differential exposure to gonadal hormones early in life. Gonadal hormones also impact within-sex variability in disordered eating, helping to explain why not all women develop an eating disorder, and some men do. We review recent findings regarding these gonadal hormone effects and their implications for the etiology of eating disorders. RECENT FINDINGS: Males are exposed to significantly higher testosterone levels than females perinatally, and this exposure appears to protect against later binge eating in males relative to females. Within-sex, higher estradiol levels among females and higher testosterone levels among males appear to be protective. Progesterone exhibits minimal direct phenotypic effects on disordered eating but appears to counteract the protective effects of estrogen in adult females. Importantly, gonadal hormone effects may be moderated by psychosocial factors. SUMMARY: Evidence suggests that gonadal hormones play a critical role in the etiology of disordered eating. Overall, higher testosterone and estrogen appear to be protective across development. Additional research is needed to identify mechanisms underlying these effects and further explore interactions between hormonal and psychosocial risk.


Assuntos
Transtornos da Alimentação e da Ingestão de Alimentos/fisiopatologia , Hormônios Gonadais/fisiologia , Animais , Feminino , Humanos , Individualidade , Masculino , Gravidez , Fatores Sexuais
8.
Metabolism ; 98: 84-94, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31226351

RESUMO

BACKGROUND: Kisspeptins, encoded by Kiss1, have emerged as essential regulators of puberty and reproduction by primarily acting on GnRH neurons, via their canonical receptor, Gpr54. Mounting, as yet fragmentary, evidence strongly suggests that kisspeptin signaling may also participate in the control of key aspects of body energy and metabolic homeostasis. However, characterization of such metabolic dimension of kisspeptins remains uncomplete, without an unambiguous discrimination between the primary metabolic actions of kisspeptins vs. those derived from their ability to stimulate the secretion of gonadal hormones, which have distinct metabolic actions on their own. In this work, we aimed to tease apart primary vs. secondary effects of kisspeptins in the control of key aspects of metabolic homeostasis using genetic models of impaired kisspeptin signaling and/or gonadal hormone status. METHODS: Body weight (BW) gain and composition, food intake and key metabolic parameters, including glucose tolerance, were comparatively analyzed, in lean and obesogenic conditions, in mice lacking kisspeptin signaling due to global inactivation of Gpr54 (displaying profound hypogonadism; Gpr54-/-) vs. Gpr54 null mice with selective re-introduction of Gpr54 expression only in GnRH cells (Gpr54-/-Tg), where kisspeptin signaling elsewhere than in GnRH neurons is ablated but gonadal function is preserved. RESULTS: In male mice, global elimination of kisspeptin signaling resulted in decreased BW, feeding suppression and increased adiposity, without overt changes in glucose tolerance, whereas Gpr54-/- female mice displayed enhanced BW gain at adulthood, increased adiposity and perturbed glucose tolerance, despite reduced food intake. Gpr54-/-Tg rescued mice showed altered postnatal BW gain in males and mildly perturbed glucose tolerance in females, with intermediate phenotypes between control and global KO animals. Yet, body composition and leptin levels were similar to controls in gonadal-rescued mice. Exposure to obesogenic insults, such as high fat diet (HFD), resulted in exaggerated BW gain and adiposity in global Gpr54-/- mice of both sexes, and worsening of glucose tolerance, especially in females. Yet, while rescued Gpr54-/-Tg males displayed intermediate BW gain and feeding profiles and impaired glucose tolerance, rescued Gpr54-/-Tg females behaved as controls, except for a modest deterioration of glucose tolerance after ovariectomy. CONCLUSION: Our data support a global role of kisspeptin signaling in the control of body weight and metabolic homeostasis, with a dominant contribution of gonadal hormone-dependent actions. However, our results document also discernible primary effects of kisspeptin signaling in the regulation of body weight gain, feeding and responses to obesogenic insults, which occur in a sexually-dimorphic manner. SUMMARY OF TRANSLATIONAL RELEVANCE: Kisspeptins, master regulators of reproduction, may also participate in the control of key aspects of body energy and metabolic homeostasis; yet, the nature of such metabolic actions remains debatable, due in part to the fact that kisspeptins modulate gonadal hormones, which have metabolic actions on their own. By comparing the metabolic profiles of two mouse models with genetic inactivation of kisspeptin signaling but different gonadal status (hypogonadal vs. preserved gonadal function), we provide herein a systematic dissection of gonadal-dependent vs. -independent metabolic actions of kisspeptins. Our data support a global role of kisspeptin signaling in the control of body weight and metabolic homeostasis, with a dominant contribution of gonadal hormone-dependent actions. However, our results document also discernible primary effects of kisspeptin signaling in the regulation of body weight gain, feeding and responses to obesogenic insults, which occur in a sexually-dimorphic manner. These data pave the way for future analyses addressing the eventual contribution of altered kisspeptin signaling in the development of metabolic alterations, especially in conditions linked to reproductive dysfunction.


Assuntos
Peso Corporal/fisiologia , Hormônios Gonadais/fisiologia , Homeostase/fisiologia , Kisspeptinas/fisiologia , Transdução de Sinais/fisiologia , Animais , Dieta , Ingestão de Alimentos , Feminino , Intolerância à Glucose/genética , Masculino , Camundongos , Camundongos Knockout , Obesidade/genética , Ovariectomia , Receptores de Kisspeptina-1/genética , Receptores de Kisspeptina-1/metabolismo , Aumento de Peso/genética
9.
Horm Behav ; 114: 104547, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31228420

RESUMO

Dehydration impairs cognitive performance in humans and rodents, although studies in animal models are limited. Estrogens have both protective effects on fluid regulation and improve performance in certain cognitive tasks. We, therefore, tested whether sex and gonadal hormones influence object recognition memory during dehydration. Because past studies used fluid deprivation to induce dehydration, which is a mixture of intracellular and extracellular fluid loss, we tested the effects of osmotic (loss of intracellular fluid) and hypovolemic (loss of extracellular fluid) dehydration on object recognition memory. After training trials consisting of exposure to two identical objects, rats were either treated with hypertonic saline to induce osmotic dehydration, furosemide to induce hypovolemic dehydration, or received a control injection and then object recognition memory was tested by presenting the original and a novel object. After osmotic dehydration, regardless of group or treatment, all rats spent significantly more time investigating the novel object. After hypovolemic dehydration, regardless of treatment, both the males and estrous females spent significantly more time investigating the novel object. While the control-treated diestrous females also spent significantly more time investigating the novel object, the furosemide-treated diestrous females spent a similar amount of time investigating the novel and original object. Follow up studies determined that loss of ovarian hormones after ovariectomy, but not loss of testicular hormones after castration, resulted in impaired memory performance in the object recognition test after hypovolemic dehydration. This series of experiments provides evidence for a protective role of ovarian hormones on dehydration-induced memory impairments.


Assuntos
Desidratação/complicações , Hormônios Gonadais/fisiologia , Transtornos da Memória/etiologia , Transtornos da Memória/prevenção & controle , Reconhecimento Psicológico/fisiologia , Animais , Desidratação/psicologia , Feminino , Hormônios Gonadais/sangue , Masculino , Transtornos da Memória/sangue , Orquiectomia , Ovariectomia , Ratos , Ratos Sprague-Dawley
10.
Behav Neurosci ; 133(5): 517-526, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31246079

RESUMO

Prior studies suggest that levels of ovarian hormones may affect learning and memory in rats, including studies of fear conditioning and extinction. We previously showed that female rats show reduced retention of extinction compared to males when measuring fear-potentiated startle, but not when measuring freezing behavior. One commonly reported observation in studies of freezing behavior is that rats with increased levels of estradiol during extinction learning show better retention of extinction than rats given extinction training when levels of estradiol are low. Here, we tested the hypothesis that fear extinction retention in a fear-potentiated startle paradigm in females is influenced by levels gonadal hormones, which we had not accounted for in our original report. We used the fear-potentiated startle paradigm to test if extinction learning was affected by estrous phase, ovariectomy, or acute systemic injections of estradiol in ovariectomized rats. We report that neither the expression nor extinction of fear-potentiated startle differed in rats given extinction training in proestrus compared to those in metestrus. Removal of the ovaries had no effect on fear acquisition or extinction learning as assessed by fear-potentiated startle. Finally, systemic injections of estradiol given to ovariectomized rats before extinction training had no effect on the expression of fear or the retention of extinction. Our findings suggest that the effect of female gonadal hormones on fear conditioning and extinction may depend on the measure of fear employed or by the parameters used to study fear learning. (PsycINFO Database Record (c) 2019 APA, all rights reserved).


Assuntos
Extinção Psicológica/efeitos dos fármacos , Medo/efeitos dos fármacos , Hormônios Gonadais/fisiologia , Animais , Estradiol/farmacologia , Estrogênios/farmacologia , Ciclo Estral/efeitos dos fármacos , Extinção Psicológica/fisiologia , Medo/fisiologia , Feminino , Hormônios Gonadais/metabolismo , Ovariectomia , Ratos , Ratos Sprague-Dawley , Reflexo de Sobressalto/fisiologia
11.
Brain Res Bull ; 149: 175-183, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31022436

RESUMO

OBJECTIVE: The role of gonadal hormones in chronic intermittent hypoxia (CIH)-evoked hypoglossal nerve (XII) neuroplasticity has not been thoroughly studied. The purpose of this study was to reveal the effects of gonadal hormone concentration variations on the XII discharge activity of rats exposed to CIH and the corresponding relationship with 5-hydroxytryptamine (5-HT). METHODS: This study employed five groups of female rats and six groups of male rats. Gonadal hormone levels were modified through gonadal resection and daily supplementation with gonadal hormones in rats of both sexes. Rats in the CIH groups were exposed to an additional 4 weeks of CIH once the operative incision for gonadectomy had healed. Finally, XII spontaneous discharge activities were recorded, and serum estradiol, testosterone and 5-HT concentrations were detected by ELISA. RESULTS: Among the female rats, the normal estradiol level groups expressed XII neuroplasticity, while the low estradiol level group failed to express this phenomenon. XII neuroplasticity was related to the serum estradiol concentration. In the male rats, XII neuroplasticity was successfully evoked in the normal testosterone level group but was suppressed in the low testosterone level group and aromatase inhibitor group. XII neuroplasticity was not significantly related to serum testosterone concentrations. Both estradiol and testosterone concentrations were related to 5-HT concentrations. CONCLUSIONS: This is the first study to analyze the effects of gonadal hormones on XII neuroplasticity in both female and male rats. The results suggest that the estradiol level is related to XII neuroplasticity rather than the testosterone level, and testosterone may indirectly adjust XII neuroplasticity by converting to estradiol. Estradiol and testosterone levels are related to 5-HT levels in the respective genders.


Assuntos
Hormônios Gonadais/fisiologia , Nervo Hipoglosso/metabolismo , Hipóxia/metabolismo , Animais , Estradiol/farmacologia , Feminino , Hormônios Gonadais/metabolismo , Hormônios Gonadais/farmacologia , Hipóxia/fisiopatologia , Masculino , Plasticidade Neuronal/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Serotonina/metabolismo , Serotonina/fisiologia , Testosterona/farmacologia
12.
Behav Brain Res ; 363: 13-22, 2019 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-30703399

RESUMO

The aim of this study was to detect differences in functional outcome after experimental subarachnoid haemorrhage (SAH) in rodents with different hormonal status. For this purpose, the endovascular perforation model was applied to four groups of Sprague-Dawley-Rats: male intact, male neutered, female intact and female neutered animals. Initial impact was measured by ICP, CPP and cerebral blood flow in the first hour after SAH. From day 4-14, the modified hole board test was applied to assess functional and neuro-cognitive outcome. Histological outcome was examined in the motor cortex and hippocampus of each hemisphere. Mortality was highest in the female intact group albeit not statistically significant. Physiologic parameters did not differ significantly between groups either. In the modified hole board test, male intact animals showed a greater impairment of declarative memory than the female intact and neutered groups. However, male intact animals showed greater avoidance behaviour and male animals revealed higher anxiety levels independent of hormonal status. No differences in histological damage of hippocampus and motor cortex between groups could be shown. We therefore speculate that the marginal deficits in cognitive performance that are shown by the male intact group in the modified hole board test are mostly caused by higher anxiety levels and cannot be interpreted as pure cognitive impairment.


Assuntos
Cognição/fisiologia , Hormônios Gonadais/fisiologia , Hemorragia Subaracnóidea/patologia , Animais , Pressão Sanguínea/fisiologia , Encéfalo/patologia , Circulação Cerebrovascular/fisiologia , Feminino , Hormônios Gonadais/metabolismo , Hipocampo/patologia , Pressão Intracraniana/fisiologia , Masculino , Memória , Testes de Estado Mental e Demência , Ratos , Ratos Sprague-Dawley , Fatores Sexuais , Hemorragia Subaracnóidea/metabolismo
13.
Horm Behav ; 108: 50-61, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30597139

RESUMO

Males and females can respond differentially to the same environmental stimuli and experimental conditions. Chronic sleep loss is a frequent and growing problem in many modern societies and has a broad variety of negative outcomes for health and well-being. While much has been done to explore the deleterious effects of sleep deprivation (SD) on cognition in both human and animal studies over the last few decades, very little attention has been paid to the part played by sex differences and gonadal steroids in respect of changes in cognitive functions caused by sleep loss. The effects of gonadal hormones on sleep regulation and cognitive performances are well established. Reduced gonadal function in menopausal women and elderly men is associated with sleep disturbances and cognitive decline as well as dementia, which suggests that sex steroids play a key role in modulating these conditions. Finding out whether there are sex differences in respect of the effect of insufficient sleep on cognition, and how neuroendocrine mediators influence cognitive impairment induced by SD could provide valuable insights into the best therapies for each sex. In this review, we aim to highlight the involvement of sex differences and gonadal hormone status on the severity of cognitive deficits induced by sleep deficiency in both human and animal studies.


Assuntos
Transtornos Cognitivos/etiologia , Hormônios Gonadais/fisiologia , Privação do Sono/complicações , Privação do Sono/psicologia , Sono/fisiologia , Idoso , Idoso de 80 Anos ou mais , Animais , Cognição/efeitos dos fármacos , Cognição/fisiologia , Transtornos Cognitivos/metabolismo , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/metabolismo , Feminino , Hormônios Gonadais/farmacologia , Hormônios Esteroides Gonadais/farmacologia , Hormônios Esteroides Gonadais/fisiologia , Humanos , Masculino , Menopausa/efeitos dos fármacos , Menopausa/fisiologia , Menopausa/psicologia , Sistemas Neurossecretores/efeitos dos fármacos , Sistemas Neurossecretores/fisiopatologia , Caracteres Sexuais , Privação do Sono/metabolismo
14.
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
15.
Compr Physiol ; 9(1): 375-411, 2018 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-30549017

RESUMO

Epidemiological studies demonstrate disparities between men and women in cardiovascular disease prevalence, clinical symptoms, treatments, and outcomes. Enrollment of women in clinical trials is lower than men, and experimental studies investigating molecular mechanisms and efficacy of certain therapeutics in cardiovascular disease have been primarily conducted in male animals. These practices bias data interpretation and limit the implication of research findings in female clinical populations. This review will focus on the biological origins of sex differences in cardiovascular physiology, health, and disease, with an emphasis on the sex hormones, estrogen and testosterone. First, we will briefly discuss epidemiological evidence of sex disparities in cardiovascular disease prevalence and clinical manifestation. Second, we will describe studies suggesting sexual dimorphism in normal cardiovascular function from fetal life to older age. Third, we will summarize and critically discuss the current literature regarding the molecular mechanisms underlying the effects of estrogens and androgens on cardiac and vascular physiology and the contribution of these hormones to sex differences in cardiovascular disease. Fourth, we will present cardiovascular disease risk factors that are positively associated with the female sex, and thus, contributing to increased cardiovascular risk in women. We conclude that inclusion of both men and women in the investigation of the role of estrogens and androgens in cardiovascular physiology will advance our understanding of the mechanisms underlying sex differences in cardiovascular disease. In addition, investigating the role of sex-specific factors in the development of cardiovascular disease will reduce sex and gender disparities in the treatment and diagnosis of cardiovascular disease. © 2019 American Physiological Society. Compr Physiol 9:375-411, 2019.


Assuntos
Doenças Cardiovasculares/epidemiologia , Hormônios Gonadais/fisiologia , Animais , Doenças Cardiovasculares/etiologia , Feminino , Humanos , Masculino , Gravidez/fisiologia , Fatores Sexuais
16.
Psychoneuroendocrinology ; 96: 173-178, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29980009

RESUMO

Cognitive impairments are a core feature of schizophrenia and contribute significantly to functional complications. Current pharmacological treatments do not ameliorate cognitive dysfunction and the aetiology of cognitive impairments are poorly understood. Hormones of the hypothalamic-pituitary-gonadal (HPG) axis that regulate reproductive function have multiple effects on the development, maintenance and function of the brain and have been suggested to also influence cognition. The aim of the current study was to investigate how HPG axis hormones effect cognition, specifically exploring the influence of menopause status and menstrual cycle irregularity on cognitive performance in women with schizophrenia. The data for the present study represents pooled baseline data from three clinical trials. Two hundred and forty female participants with a diagnosis of schizophrenia or schizoaffective disorder were included in the analysis. Cognition was assessed using the Repeatable Battery for the Assessment of Neuropsychological Status. Hormone assays for serum sex steroids and pituitary hormones (including estradiol, progesterone, luteinising hormone and follicle-stimulating hormone) were conducted and women were classified as postmenopausal; perimenopausal; premenopausal/reproductive, further classified into regular and irregular menstrual cycles. To model a comparison of cognitive performance for i) perimenopausal; ii) post-menopausal women and iii) reproductive aged women with irregular cycles to reproductive aged women with regular cycles a semiparametric regression model (generalised additive mode) was fitted. The results revealed that in females with schizophrenia, menstrual cycle irregularity predicted significantly poorer cognitive performance in the areas of psychomotor speed, verbal fluency and verbal memory. Perimenopause was not associated with cognitive changes and the post-menopausal period was associated with poorer visuospatial performance. This study provides evidence to associate reproductive hormones with cognitive dysfunction in schizophrenia.


Assuntos
Cognição/fisiologia , Ciclo Menstrual/psicologia , Distúrbios Menstruais/complicações , Adulto , Estradiol/análise , Estradiol/sangue , Feminino , Hormônio Foliculoestimulante/análise , Hormônio Foliculoestimulante/sangue , Hormônios Gonadais/fisiologia , Humanos , Hormônio Luteinizante/análise , Hormônio Luteinizante/sangue , Memória/fisiologia , Menopausa/psicologia , Ciclo Menstrual/fisiologia , Distúrbios Menstruais/fisiopatologia , Pessoa de Meia-Idade , Testes Neuropsicológicos , Perimenopausa , Pós-Menopausa , Pré-Menopausa , Progesterona/análise , Progesterona/sangue , Transtornos Psicóticos , Esquizofrenia/complicações , Esquizofrenia/fisiopatologia
17.
Biol Sex Differ ; 9(1): 32, 2018 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-30001741

RESUMO

BACKGROUND: Our previous study revealed that adult female rats respond differently to trauma than adult males, recapitulating sex differences in symptoms of post-traumatic stress disorder (PTSD) exhibited by women and men. Here, we asked two questions: does the female phenotype depend on (1) social housing condition and/or (2) circulating gonadal hormones? METHODS: For the first study, the effects of single prolonged stress (SPS) were compared for females singly or pair-housed. For the second study, adult male and female rats were gonadectomized or sham-gonadectomized 2 weeks prior to exposure to SPS, with half the gonadectomized rats given testosterone. In addition to the typical measures of the trauma response in rats, acoustic startle response (ASR), and the dexamethasone suppression test (DST), we also used two other measures typically used to assess depressive-like responses, social interaction and sucrose preference. Glucocorticoid receptor (GR) expression in the hypothalamus was also examined. RESULTS: We now report that the distinct trauma response of female rats is not influenced by social housing condition. Moreover, sex differences in the response to SPS based on ASR and DST, replicated in the current study, are independent of adult gonadal hormones. Regardless of hormonal status, traumatized males show a hyper-responsive phenotype whereas traumatized females do not. Moreover, testosterone treatment in adulthood did not masculinize the response to trauma in females. Notably, both sucrose preference and social interaction tests revealed an effect of trauma in females but not in males, with the effects of SPS on sucrose preference dependent on ovarian hormones. Effects of SPS on GR expression in the hypothalamus also depended on gonadal hormones in females. CONCLUSIONS: We propose that the trauma response for female rats is depressive in nature, recapitulating the female bias in PTSD for internalizing symptoms and major depression in contrast to the externalizing symptoms of males. Presumed core markers of PTSD (enhanced ASR and negative feedback control of corticosterone) are apparently relevant only to males and are independent of adult gonadal hormones. Such sex differences in trauma responding are likely determined earlier in life. We conclude that males and females show fundamentally different responses to trauma that do not simply reflect differences in resilience.


Assuntos
Hormônios Gonadais/fisiologia , Caracteres Sexuais , Estresse Psicológico , Anedonia , Animais , Encéfalo/metabolismo , Dexametasona/administração & dosagem , Éter/administração & dosagem , Feminino , Relações Interpessoais , Masculino , Ratos Sprague-Dawley , Receptores de Glucocorticoides/metabolismo , Reflexo de Sobressalto , Restrição Física , Estresse Fisiológico , Natação
18.
J Neurosci Res ; 96(8): 1388-1397, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29741787

RESUMO

Exposure to stress activates both the hypothalamus-pituitary-adrenal (HPA) axis and the sympathetic nervous system (SNS). A growing body of research points to the contribution of sex hormones (testosterone, estrogen, and progesterone), the end products of the hypothalamus-pituitary-gonadal (HPG) axis, in modulating stress reactivity. The present study aimed at investigating the potential modulating role of sex hormones on HPA and SNS reactivity to psychosocial stress. The reactivity, induced by the Trier Social Stress Test, was analyzed by measuring the levels of cortisol and alpha-amylase (markers for SNS activity) in four saliva samples each of 21 men and 37 women (17 not using oral contraceptives and in their luteal phase, and 20 women using oral contraceptives). In addition, basal sex hormones were sampled prior to the psychosocial stress exposure. Results revealed that controlling for testosterone, estrogen, and progesterone diminished the impact of stress on cortisol reactivity and on alpha-amylase reactivity. Moreover, controlling for sex hormones also diminished the differential pattern of cortisol reactivity in each experimental group among responders. Furthermore, correlation analyses revealed differences between groups in the association between sex hormones and alpha-amylase. The present findings indicate a modulatory role for sex hormones in HPA and SNS reactivity and emphasize the need for control of sex hormone fluctuations when examining cortisol and alpha-amylase reactivity to stress.


Assuntos
Hormônios Gonadais/fisiologia , Sistema Hipotálamo-Hipofisário/fisiologia , Sistema Hipófise-Suprarrenal/fisiologia , Estresse Psicológico/psicologia , Sistema Nervoso Simpático/fisiologia , Adulto , Biomarcadores/metabolismo , Anticoncepcionais Orais , Teste de Esforço , Feminino , Humanos , Masculino , Saliva/metabolismo
19.
Am J Physiol Heart Circ Physiol ; 315(2): H366-H374, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29727219

RESUMO

Sex hormone concentrations differ between men, premenopausal women with natural menstrual cycles (NAT), and premenopausal women using oral contraceptive pills (OCP), as well as across menstrual or OCP phases. This study sought to investigate how differences in sex hormones, particularly estradiol, between men and women and across cycle phases might influence brachial artery endothelial function. Fifty-three healthy adults (22 ± 3 yr, 20 men, 15 NAT women, and 18 second-, third-, or fourth-generation OCP women) underwent assessments of sex hormones and endothelial [flow-mediated dilation (FMD) test] and smooth muscle [nitroglycerin (NTG) test] function. Men were tested three times at 1-wk intervals, and women were tested three times throughout a single menstrual or OCP cycle (NAT: menstrual, midfollicular, and luteal phases and OCP: placebo/no pill, "early", and "late" active pill phases). Endogenous estradiol concentration was comparable between men and women in their NAT menstrual or OCP placebo phase ( P = 0.36) but increased throughout a NAT cycle ( P < 0.001). Allometrically scaled FMD did not change across a NAT or OCP cycle but was lower in both groups of women than in men ( P = 0.005), whereas scaled NTG was lower only in NAT women ( P = 0.001). Changes in estradiol across a NAT cycle were not associated with changes in relative FMD ( r2 = 0.01, P = 0.62) or NTG ( r2 = 0.09, P = 0.13). Duration of OCP use was negatively associated with the average relative FMD for second-generation OCP users only ( r = -0.65, P = 0.04). Our findings suggest that brachial endothelial function is unaffected by cyclic hormonal changes in premenopausal women but may be negatively impacted by longer-term use of second-generation OCPs. NEW & NOTEWORTHY We demonstrate that brachial artery flow-mediated dilation does not change across a menstrual or oral contraceptive pill cycle in premenopausal women but is lower in women than in men. Although unaffected by within-cycle changes in estradiol, brachial flow-mediated dilation is negatively correlated with duration of oral contraceptive pill use for second-generation pills.


Assuntos
Artéria Braquial/fisiologia , Anticoncepcionais Orais Hormonais/farmacologia , Endotélio Vascular/fisiologia , Ciclo Menstrual/fisiologia , Adulto , Velocidade do Fluxo Sanguíneo , Artéria Braquial/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Feminino , Hormônios Gonadais/fisiologia , Humanos , Masculino , Distribuição Aleatória
20.
Am J Physiol Heart Circ Physiol ; 315(2): H357-H365, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29677465

RESUMO

Arterial stiffness is associated with increased cardiovascular disease risk. Previous sex-based investigations of local and central stiffness report inconsistent findings and have not controlled for menstrual cycle phase in women. There is also evidence that sex hormones influence the vasculature, but their impact on arterial stiffness across a natural menstrual (NAT) or oral contraceptive pill (OCP) cycle has been understudied. This study sought to 1) examine potential sex differences in local and central stiffness, 2) compare stiffness profiles between NAT and OCP cycles, and 3) investigate the relationship between duration of OCP use and arterial stiffness. Sex hormone concentrations, ß-stiffness index (local stiffness), and carotid-femoral pulse wave velocity [cfPWV (central stiffness)] were assessed in 53 healthy adults (22 ± 3 yr old, 20 men, 15 NAT women, and 18 OCP women). All participants were tested three times: men on the same day and time 1 wk apart, NAT women in menstrual, midfollicular and luteal phases of the menstrual cycle, and OCP women in placebo, early active and late active pill phases. ß-Stiffness was higher in men than NAT and OCP women ( P < 0.001), whereas cfPWV was similar between groups ( P = 0.09). ß-Stiffness and cfPWV did not differ across or between NAT and OCP cycles ( P > 0.05 for both) and were not associated with duration of OCP use (ß-stiffness: r = 0.003, P = 0.99; cfPWV: r = -0.26, P = 0.30). The apparent sex differences in local, but not central, stiffness highlight the importance of assessing both indexes in comparisons between men and women. Furthermore, fluctuating sex hormone levels do not appear to influence ß-stiffness or cfPWV. Therefore, these stiffness indexes may need to be assessed during only one cycle phase in women in future investigations. NEW & NOTEWORTHY We observed higher local, but not central, arterial stiffness in men than women. We also demonstrated that there are no differences in arterial stiffness between naturally cycling women and women who use monophasic oral contraceptive pills, and that the duration of oral contraceptive pill use does not influence arterial stiffness.


Assuntos
Anticoncepcionais Orais Hormonais/farmacologia , Hormônios Gonadais/fisiologia , Ciclo Menstrual/fisiologia , Rigidez Vascular/efeitos dos fármacos , Adolescente , Adulto , Artérias Carótidas/fisiologia , Feminino , Artéria Femoral/fisiologia , Humanos , Masculino , Análise de Onda de Pulso , Rigidez Vascular/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...