Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.149
Filtrar
1.
CNS Neurosci Ther ; 30(5): e14726, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38715251

RESUMO

AIMS: The preoptic area (POA) of the hypothalamus, crucial in thermoregulation, has long been implicated in the pain process. However, whether nociceptive stimulation affects body temperature and its mechanism remains poorly studied. METHODS: We used capsaicin, formalin, and surgery to induce acute nociceptive stimulation and monitored rectal temperature. Optical fiber recording, chemical genetics, confocal imaging, and pharmacology assays were employed to confirm the role and interaction of POA astrocytes and extracellular adenosine. Immunofluorescence was utilized for further validation. RESULTS: Acute nociception could activate POA astrocytes and induce a decrease in body temperature. Manipulation of astrocytes allowed bidirectional control of body temperature. Furthermore, acute nociception and astrocyte activation led to increased extracellular adenosine concentration within the POA. Activation of adenosine A1 or A2A receptors contributed to decreased body temperature, while inhibition of these receptors mitigated the thermo-lowering effect of astrocytes. CONCLUSION: Our results elucidate the interplay between acute nociception and thermoregulation, specifically highlighting POA astrocyte activation. This enriches our understanding of physiological responses to painful stimuli and contributes to the analysis of the anatomical basis involved in the process.


Assuntos
Astrócitos , Hipotermia , Nociceptividade , Área Pré-Óptica , Animais , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/metabolismo , Astrócitos/metabolismo , Astrócitos/efeitos dos fármacos , Nociceptividade/fisiologia , Hipotermia/induzido quimicamente , Masculino , Camundongos , Receptores Purinérgicos P1/metabolismo , Camundongos Endogâmicos C57BL , Adenosina/metabolismo , Capsaicina/farmacologia , Formaldeído/toxicidade , Formaldeído/farmacologia
2.
Mol Metab ; 84: 101951, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38729241

RESUMO

OBJECTIVE: Hypothalamic signals potently stimulate energy expenditure by engaging peripheral mechanisms to restore energy homeostasis. Previous studies have identified several critical hypothalamic sites (e.g. preoptic area (POA) and ventromedial hypothalamic nucleus (VMN)) that could be part of an interconnected neurocircuit that controls tissue thermogenesis and essential for body weight control. However, the key neurocircuit that can stimulate energy expenditure has not yet been established. METHODS: Here, we investigated the downstream mechanisms by which VMN neurons stimulate adipose tissue thermogenesis. We manipulated subsets of VMN neurons acutely as well as chronically and studied its effect on tissue thermogenesis and body weight control, using Sf1Cre and Adcyap1Cre mice and measured physiological parameters under both high-fat diet and standard chow diet conditions. To determine the node efferent to these VMN neurons, that is involved in modulating energy expenditure, we employed electrophysiology and optogenetics experiments combined with measurements using tissue-implantable temperature microchips. RESULTS: Activation of the VMN neurons that express the steroidogenic factor 1 (Sf1; VMNSf1 neurons) reduced body weight, adiposity and increased energy expenditure in diet-induced obese mice. This function is likely mediated, at least in part, by the release of the pituitary adenylate cyclase-activating polypeptide (PACAP; encoded by the Adcyap1 gene) by the VMN neurons, since we previously demonstrated that PACAP, at the VMN, plays a key role in energy expenditure control. Thus, we then shifted focus to the subpopulation of VMNSf1 neurons that contain the neuropeptide PACAP (VMNPACAP neurons). Since the VMN neurons do not directly project to the peripheral tissues, we traced the location of the VMNPACAP neurons' efferents. We identified that VMNPACAP neurons project to and activate neurons in the caudal regions of the POA whereby these projections stimulate tissue thermogenesis in brown and beige adipose tissue. We demonstrated that selective activation of caudal POA projections from VMNPACAP neurons induces tissue thermogenesis, most potently in negative energy balance and activating these projections lead to some similar, but mostly unique, patterns of gene expression in brown and beige tissue. Finally, we demonstrated that the activation of the VMNPACAP neurons' efferents that lie at the caudal POA are necessary for inducing tissue thermogenesis in brown and beige adipose tissue. CONCLUSIONS: These data indicate that VMNPACAP connections with the caudal POA neurons impact adipose tissue function and are important for induction of tissue thermogenesis. Our data suggests that the VMNPACAP → caudal POA neurocircuit and its components are critical for controlling energy balance by activating energy expenditure and body weight control.


Assuntos
Metabolismo Energético , Neurônios , Área Pré-Óptica , Termogênese , Núcleo Hipotalâmico Ventromedial , Animais , Núcleo Hipotalâmico Ventromedial/metabolismo , Termogênese/fisiologia , Área Pré-Óptica/metabolismo , Camundongos , Neurônios/metabolismo , Masculino , Fator Esteroidogênico 1/metabolismo , Fator Esteroidogênico 1/genética , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Dieta Hiperlipídica , Camundongos Endogâmicos C57BL , Peso Corporal , Tecido Adiposo Marrom/metabolismo
3.
Behav Brain Res ; 465: 114928, 2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38432301

RESUMO

Testosterone (T), estrogen receptor alpha (ERα), and androgen receptor (AR) play a significant role in the regulation of paternal behavior. We determined the effects of deprivation of paternal care on alterations in paternal behavior, T concentrations in plasma, and the presence of ERα and AR in the medial preoptic area (mPOA), bed nucleus of the stria terminalis (BNST), medial amygdala (MeA), and olfactory bulb (OB), as well as the corticosterone (CORT) concentrations in plasma caused by deprivation of paternal care in the Mongolian gerbil (Meriones unguiculatus). Twenty pairs of gerbils were formed; the pups were deprived of paternal care (DPC) in 10 pairs. In another 10 pairs, the pups received paternal care (PC). Ten males raised in DPC condition and 10 males raised in PC conditions were mated with virgin females. When they became fathers, each DPC male and PC male was subjected to tests of paternal behavior on day three postpartum. Blood samples were obtained to quantify T and CORT concentrations, and the brains were removed for ERα and AR immunohistochemistry analyses. DPC males gave less care to their pups than PC males, and they had significantly lower T concentrations and levels of ERα and AR in the mPOA and BNST than PC males. DPC males also had higher CORT concentrations than PC males. These results suggest that in the Mongolian gerbil father's absence causes a decrease in paternal care in the offspring, which is associated with alterations in the neuroendocrine mechanisms that regulate it.


Assuntos
Receptores Androgênicos , Núcleos Septais , Animais , Feminino , Masculino , Humanos , Gerbillinae/fisiologia , Receptores Androgênicos/metabolismo , Núcleos Septais/metabolismo , Receptor alfa de Estrogênio/metabolismo , Comportamento Paterno/fisiologia , Área Pré-Óptica/metabolismo , Pai , Corticosterona
4.
Behav Brain Res ; 465: 114965, 2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38522595

RESUMO

Brain areas important for social perception, social reward, and social behavior - collectively referred to as the social-decision-making network (SDN) - appear to be highly conserved across taxa. These brain areas facilitate a variety of social behaviors such as conspecific approach/avoidance, aggression, mating, parental care, and recognition. Although the SDN has been investigated across taxa, little is known about its functioning in reptiles. Research on the snake SDN may provide important new insights, as snakes have a keen social perceptual system and express a relatively reduced repertoire of social behaviors. Here, we present the results of an experiment in which ball pythons (Python regius) interacted with a same-sex conspecific for one hour and neural activation was investigated through Fos immunoreactivity. Compared to controls, snakes that interacted socially had higher Fos counts in brain areas implicated in social behavior across taxa, such as the medial amygdala, preoptic area, nucleus accumbens, and basolateral amygdala. Additionally, we found differential Fos immunoreactivity in the ventral amygdala, which facilitates communication between social brain areas. In many of these areas, Fos counts differed by sex, which may be due to increased competition between males. Fos counts did not differ in early sensory (i.e., vomeronasal) processing structures. As ball python social systems lack parental care, cooperation, or long-term group living, these results provide valuable insight into the basal functions of the vertebrate social decision-making network.


Assuntos
Encéfalo , Proteínas Proto-Oncogênicas c-fos , Masculino , Animais , Proteínas Proto-Oncogênicas c-fos/metabolismo , Encéfalo/metabolismo , Área Pré-Óptica/metabolismo , Núcleo Accumbens/metabolismo , Serpentes/metabolismo
5.
Front Endocrinol (Lausanne) ; 14: 1212854, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37900129

RESUMO

Background: The neuroendocrine control of ovulation is orchestrated by neuronal circuits that ultimately drive the release of gonadotropin-releasing hormone (GnRH) from the hypothalamus to trigger the preovulatory surge in luteinizing hormone (LH) secretion. While estrogen feedback signals are determinant in triggering activation of GnRH neurons, through stimulation of afferent kisspeptin neurons in the rostral periventricular area of the third ventricle (RP3VKISS1 neurons), many neuropeptidergic and classical neurotransmitter systems have been shown to regulate the LH surge. Among these, several lines of evidence indicate that the monoamine neurotransmitter serotonin (5-HT) has an excitatory, permissive, influence over the generation of the surge, via activation of type 2 5-HT (5-HT2) receptors. The mechanisms through which this occurs, however, are not well understood. We hypothesized that 5-HT exerts its influence on the surge by stimulating RP3VKISS1 neurons in a 5-HT2 receptor-dependent manner. Methods: We tested this using kisspeptin neuron-specific calcium imaging and electrophysiology in brain slices obtained from male and female mice. Results: We show that exogenous 5-HT reversibly increases the activity of the majority of RP3VKISS1 neurons. This effect is more prominent in females than in males, is likely mediated directly at RP3VKISS1 neurons and requires activation of 5-HT2 receptors. The functional impact of 5-HT on RP3VKISS1 neurons, however, does not significantly vary during the estrous cycle. Conclusion: Taken together, these data suggest that 5-HT2 receptor-mediated stimulation of RP3VKISS1 neuron activity might be involved in mediating the influence of 5-HT on the preovulatory LH surge.


Assuntos
Kisspeptinas , Área Pré-Óptica , Camundongos , Feminino , Masculino , Animais , Área Pré-Óptica/metabolismo , Kisspeptinas/metabolismo , Serotonina/farmacologia , Neurônios/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Receptores de Serotonina , Neurotransmissores
6.
Ann Anat ; 250: 152132, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37454827

RESUMO

BACKGROUND: Decreased estrogen levels can cause abnormal thermosensitivity of the preoptic area (POA) in the hypothalamus during menopause, which may cause hot flashes. Thermosensitive transient receptors (ThermoTRPs) affect the thermosensitivity of neurons. It is worth exploring whether ThermoTRPs change under low estrogen state and participate in the abnormal thermoregulation of POA. METHODS: Adult female Sprague-Dawley rats were randomly divided into sham operation (SHAM), ovariectomy (OVX) and estrogen treatment after ovariectomy (OVX+E) groups. Under 10 â„ƒ, 18 â„ƒ, 25 â„ƒ, 37 â„ƒ and 45 â„ƒ incubations, their skin temperature was monitored and the expression of TRPA1, TRPM8, TRPM2, and TRPV1 in POA were investigated. RESULTS: The skin temperature of ovariectomized rats changed faster and more dramatically under different incubation temperatures. The results at mRNA level show that only the expression of TRPM2 decreased in POA of OVX group compared with the other two groups at 25 â„ƒ, TRPA1 expression in POA of the three groups increased at 10 â„ƒ, TRPM8 increased at 10 â„ƒ and 18 â„ƒ, TRPV1 increased at 10 â„ƒ and 45 â„ƒ, while the expression of TRPM2 decreased at 10 â„ƒ and 18 â„ƒ and increased at 37 â„ƒ and 45 â„ƒ. In all these cases, the magnitudes of the changes were less in the OVX group relative to the other two groups. The further immunohistochemical and Western blot results of TRPM2 and the activated TRPM2 positive cells labeled by c-Fos were consistent with the results of mRNA level. CONCLUSIONS: The expression and thermosensitivity of TRPM2 in POA changed greatly under different incubation temperatures, but the changes in ovariectomized rats were less. This may be the key factor triggering thermoregulation dysfunction under low estrogen and may cause hot flashes.


Assuntos
Canais de Cátion TRPM , Canais de Potencial de Receptor Transitório , Ratos , Feminino , Animais , Humanos , Área Pré-Óptica/metabolismo , Fogachos , Ratos Sprague-Dawley , Canais de Potencial de Receptor Transitório/metabolismo , Canais de Cátion TRPM/genética , Canais de Cátion TRPM/metabolismo , Estradiol , Hipotálamo/metabolismo , Menopausa , Estrogênios , Regulação da Temperatura Corporal , RNA Mensageiro/metabolismo , Ovariectomia
7.
Physiol Behav ; 268: 114237, 2023 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-37192686

RESUMO

Two distinct estrogen receptors (ERs) exist, ERα and ERß. Both receptors participate in sexual differentiation of the rat brain and likely participate in the regulation of adult sexual orientation (i.e. partner preference). This last idea was investigated herein by examining males treated with the aromatase inhibitor, letrozole, administered prenatally (0.56 µg/kg G10-22). This treatment usually provokes same-sex preference in 1-2 males per litter. Vehicle-treated males (with female preference) and females in spontaneous proestrus (with male preference) were included as controls. ERα and ERß expression was analyzed by immunohistochemistry in brain areas known to control masculine sexual behavior and partner preference, like the medial preoptic area (MPOA), bed nucleus of the stria terminalis (BNST), medial amygdala (MeA) and ventromedial hypothalamic nucleus (VMH), as well as other brain regions suspected to participate in these processes. In addition, serum levels of estradiol were determined in all male groups. Letrozole-treated male rats that preferred sexually experienced males (LPM) showed over-expressed ERα in the hippocampal cornu Ammonis (CA 1, 3, 4) and dentate gyrus. The LPM group showed up-regulated ERß expression in the CA2 and reticular thalamic nucleus. The levels of estradiol did not differ between the groups. Higher expression of ERs in these males was different than their expression in females, with male sex-preference. This suggests that males with same-sex preference showed a unique brain, this sui generis steroid receptor expression probably participates in the biological underpinnings of sexual preference.


Assuntos
Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Ratos , Animais , Feminino , Masculino , Humanos , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Letrozol/metabolismo , Receptores de Estrogênio/metabolismo , Encéfalo/metabolismo , Área Pré-Óptica/metabolismo , Comportamento Sexual , Estradiol/farmacologia , Estradiol/metabolismo
8.
Zoolog Sci ; 40(2): 128-140, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37042692

RESUMO

Many vertebrate species show breeding periods and exhibit series of characteristic species-specific sexual behaviors only during the breeding period. Here, secretion of gonadal sex hormones from the mature gonads has been considered to facilitate sexual behaviors. Thus, the sexual behavior has long been considered to be regulated by neural and hormonal mechanisms. In this review, we discuss recent progress in the study of neural control mechanisms of sexual behavior with a focus on studies using fish, which have often been the favorite animals used by many researchers who study instinctive animal behaviors. We first discuss control mechanisms of sexual behaviors by sex steroids in relation to the anatomical studies of sex steroid-concentrating neurons in various vertebrate brains, which are abundantly distributed in evolutionarily conserved areas such as preoptic area (POA) and anterior hypothalamus. We then focus on another brain area called the ventral telencephalic area, which has also been suggested to contain sex steroid-concentrating neurons and has been implicated in the control of sexual behaviors, especially in teleosts. We also discuss control of sex-specific behaviors and sexual preference influenced by estrogenic signals or by olfactory/pheromonal signals. Finally, we briefly summarize research on the modulatory control of motivation for sexual behaviors by a group of peptidergic neurons called terminal nerve gonadotropin-releasing hormone (TN-GnRH) neurons, which are known to be especially developed in fishes among various vertebrate species.


Assuntos
Peixes , Área Pré-Óptica , Feminino , Masculino , Animais , Peixes/fisiologia , Área Pré-Óptica/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/fisiologia , Encéfalo/metabolismo , Esteroides
9.
Neurosci Res ; 194: 36-43, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37030575

RESUMO

In many mammalian species, females exhibit higher sociability and gregariousness than males, presumably due to the benefit of group living for maternal care. We have previously reported that adult female mice exhibit contact-seeking behaviors upon acute social isolation via amylin-calcitonin receptor (Calcr) signaling in the medial preoptic area (MPOA). In this study, we examined the sex differences in the behavioral responses to acute social isolation and reunion, and the levels of amylin and Calcr expression in the MPOA. We found that male mice exhibited significantly less contact-seeking upon social isolation. Upon reunion, male mice contacted each other to a similar extent as females, but their interactions were more aggressive and less affiliative compared with females. While Calcr-expressing neurons were activated during social contacts in males as in females, the amylin and Calcr expression were significantly lower in males than in females. Together with our previous findings, these findings suggested that the lower expression of both amylin and Calcr may explain the lower contact-seeking and social affiliation of male mice.


Assuntos
Polipeptídeo Amiloide das Ilhotas Pancreáticas , Área Pré-Óptica , Camundongos , Animais , Feminino , Masculino , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Área Pré-Óptica/metabolismo , Isolamento Social , Caracteres Sexuais , Mamíferos
10.
J Chem Neuroanat ; 129: 102241, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36738851

RESUMO

The amylin and the melanin-concentrating hormone [MCH] are two peptides related to energetic homeostasis. During lactation, it is possible to locate neurons expressing these peptides in the preoptic area of rat dams. In addition, it was demonstrated that the number of MCH neurons in this region is modulated by litter size. Taken together, the aims of this work were (1) to verify the time course of amylin immunoreactivity during lactation; (2) to verify whether litter size modulates the number of amylin-ir neurons (3) to verify whether there is colocalization between the amylin-ir and MCH-ir neurons. Our results show that (1) there is an increase in the number of amylin-ir neurons during lactation, which reaches a peak at postpartum day 19 and drastically reduces after weaning; (2) there is no correlation between litter size and the number of amylin-ir neurons; and (3) there is minimal overlap between amylin-ir and MCH-ir neurons.


Assuntos
Hormônios Hipotalâmicos , Área Pré-Óptica , Feminino , Ratos , Animais , Área Pré-Óptica/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas , Hormônios Hipofisários , Hormônios Hipotalâmicos/metabolismo , Melaninas , Lactação , Neurônios/metabolismo
11.
Mol Brain ; 16(1): 10, 2023 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-36658598

RESUMO

Social animals become stressed upon social isolation, proactively engaging in affiliative contacts among conspecifics after resocialization. We have previously reported that calcitonin receptor (Calcr) expressing neurons in the central part of the medial preoptic area (cMPOA) mediate contact-seeking behaviors in female mice. Calcr neurons in the posterodorsal part of the medial amygdala (MeApd) are also activated by resocialization, however their role in social affiliation is still unclear. Here we first investigated the functional characteristics of MeApd Calcr + cells; these neurons are GABAergic and show female-biased Calcr expression. Next, using an adeno-associated virus vector expressing a short hairpin RNA targeting Calcr we aimed to identify its molecular role in the MeApd. Inhibiting Calcr expression in the MeApd increased social contacts during resocialization without affecting locomotor activity, suggesting that the endogenous Calcr signaling in the MeApd suppresses social contacts. These results demonstrate the distinct roles of Calcr in the cMPOA and MeApd for regulating social affiliation.


Assuntos
Complexo Nuclear Corticomedial , Receptores da Calcitonina , Feminino , Animais , Camundongos , Receptores da Calcitonina/metabolismo , Tonsila do Cerebelo/metabolismo , Neurônios/metabolismo , Área Pré-Óptica/metabolismo
12.
Horm Behav ; 148: 105296, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36528006

RESUMO

The medial preoptic area (mPOA) in the hypothalamus is an important integrator of neuroendocrine signaling and a key regulator of both natural and drug-induced reward. Although the mPOA modulates sex differences in other behaviors, whether it also modulates sex differences in cocaine response remains unclear. To help us better understand the mPOA's role in sex differences associated with cocaine response, we examined cocaine-induced changes in locomotion and neural activity in the mPOA of male and female rats. In addition, neural activity in the striatum, a brain area known to be involved in cocaine response, was examined for comparison purposes. Fos, the protein product of the immediate early gene c-fos, was used as the marker of neural activity. Locomotion chambers were used to measure behavior, radioimmunoassays and vaginal lavages were used to determine hormonal status, and immunohistochemical assays were used to quantify Fos. To account for the effects of gonadal hormones, rats were left gonadally intact and categorized as either 'low-estradiol' or 'high-estradiol' based on their hormonal status on test day. Results indicate that high-estradiol females experienced greater cocaine-induced mPOA Fos-immunoreactivity (Fos-ir) and displayed greater cocaine-induced locomotion than low estradiol females. Conversely, high-estradiol males experienced less cocaine-induced mPOA Fos-ir and displayed less cocaine-induced locomotion than low-estradiol males. Cocaine-induced Fos-ir in the mPOA also correlated with cocaine-induced Fos-ir in areas of the striatum already associated with cocaine response. These findings further support the mPOA's role in the endocrine-mediated response to cocaine. It also identifies the mPOA as a contributor to sex differences in cocaine response and potential differences in vulnerability to developing cocaine use disorders.


Assuntos
Cocaína , Estradiol , Ratos , Feminino , Masculino , Animais , Estradiol/farmacologia , Estradiol/metabolismo , Área Pré-Óptica/metabolismo , Cocaína/farmacologia , Hipotálamo/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo
13.
Behav Brain Res ; 437: 114153, 2023 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-36220415

RESUMO

According to approach-avoidance model, virgin female laboratory rats display maternal behaviour when the tendency to approach and interact with the pup is stronger than avoiding it. A positive neural mechanism that includes the medial preoptic area (mPOA)/bed nucleus of the stria terminalis (BNST) and a negative mechanism that involves the anterior hypothalamic nucleus (AHN)/ventromedial nucleus (VMN)/ periaqueductal grey (PAG) underlie to these behaviours. Unlike virgin rats, which avoid the pups, virgin females Mongolian gerbils (Meriones unguiculatus) can be immediately either maternal or aggressive with the pups. Furthermore, the Mongolian gerbil is monogamous and biparental species. Despite these difference, we hypothesised that maternal and aggressive interaction with the pups could activate mPOA/BNST and AHN/VMH/PAG, respectively, and that maternal response could be associated with high concentrations of estradiol (E2). Twenty virgin maternal females and 20 aggressive toward the pups were selected. Ten maternal females interacted with the pups (MAT-pups) and 10 with candy (MAT-candy). Of the 20 aggressive females, 10 interacted with the pups (AGG-pups) and 10 with candy (AGG-candy). Immediately after the test, blood samples were taken to quantify E2. The brains were dissected for c-Fos immunohistochemistry. MAT-pups females had significantly higher activation in mPOA/BNST than MAT-candy females, while AGG-pups showed significant activation in AHN/VMH/PAG compared with AGG-candy females. The maternal response was associated with high concentrations of E2. These results suggested a positive and a negative mechanism in the regulation of maternal behaviour in the Mongolian gerbil, and that the immediate maternal response could be due to high E2 concentrations.


Assuntos
Área Pré-Óptica , Núcleos Septais , Animais , Feminino , Ratos , Humanos , Gerbillinae , Área Pré-Óptica/metabolismo , Núcleos Septais/metabolismo , Comportamento Materno/fisiologia , Proteínas Proto-Oncogênicas c-fos/metabolismo
14.
Obesity (Silver Spring) ; 30(11): 2242-2255, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36321273

RESUMO

OBJECTIVE: Elevation of energy expenditure through an increase of brown adipose tissue (BAT) thermogenesis is regarded as one of the most promising ways to prevent obesity development. The preoptic area (POA) of the hypothalamus is a critical area for control of BAT thermogenesis. However, the intracellular signaling cascades in the POA for regulation of BAT thermogenesis are poorly understood. METHODS: Phosphorylation proteomics (phosphoproteomics) and bioinformatics approaches were used to disclose numerous hypothalamic signaling pathways involved in the regulation of BAT thermogenesis. Conditional manipulation of the p38α gene in mouse POA was performed by stereotaxic injection of adeno-associated virus 9 vector to explore the role of p38α in BAT thermogenesis. RESULTS: Multiple hypothalamic signaling pathways were triggered by cold exposure, especially the mitogen-activated protein kinase (MAPK) signaling pathway. The p38α activation, but not extracellular signal-regulated kinase 1/2 (ERK1/2) and c-Jun NH2-terminal kinase (JNK), in the hypothalamus was significantly decreased during cold exposure. p38α deficiency in the POA dramatically elevated energy expenditure owing to a marked increase in BAT thermogenesis, resulting in significantly decreased body weight gain and fat mass. Overexpression of p38α in the POA led to a dramatic increase in weight gain. CONCLUSIONS: These results demonstrate that p38α in the POA exacerbates obesity development, at least in part owing to a decrease in BAT thermogenesis.


Assuntos
Tecido Adiposo Marrom , Área Pré-Óptica , Camundongos , Animais , Tecido Adiposo Marrom/metabolismo , Área Pré-Óptica/metabolismo , Termogênese/fisiologia , Obesidade/metabolismo , Metabolismo Energético/fisiologia , Aumento de Peso
15.
J Neuroendocrinol ; 34(6): e13127, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35394094

RESUMO

In male Japanese quail, brain aromatase is crucial for the hormonal activation of sexual behaviour, but the sites producing neuro-oestrogens that are critical for these behaviours have not been completely identified. This study examined the function of aromatase expressed in several nuclei of the social behaviour network on a measure of sexual motivation known as the frequency of rhythmic cloacal sphincter movements (RCSM) and on copulatory behaviour. Sexually experienced castrated males chronically treated with testosterone were stereotaxically implanted with the aromatase inhibitor vorozole (VOR), or cholesterol as control, and tested for sexual behaviour. In experiment 1, males were implanted in the medial preoptic nucleus (POM) with VOR, a manipulation known to reduce the expression of copulatory behaviour. This experiment served as positive control, but also showed that VOR implanted in the dorsomedial or lateral portions of the POM similarly inhibits male copulatory behaviour compared to control implants. In experiments 2 to 4, males received stereotaxic implants of VOR in the periaqueductal gray (PAG), the nucleus taeniae of the amygdala (TnA) and the ventromedial nucleus of the hypothalamus (VMN), respectively. Sexual behaviour was affected only in individuals where VOR was implanted in the PAG: these males displayed significantly lower frequencies of cloacal contact movements, the last step of the copulatory sequence. Inhibition of aromatase in the TnA and VMN did not alter copulatory ability. Overall, RCSM frequency remained unaffected by VOR regardless of implantation site. Together, these results suggest that neuro-oestrogens produced in the POM contribute the most to the control of male copulatory behaviour, while aromatase expressed in the PAG might also participate to premotor aspects of male copulatory behaviour.


Assuntos
Aromatase , Coturnix , Comportamento Sexual Animal , Comportamento Social , Animais , Aromatase/metabolismo , Encéfalo/metabolismo , Coturnix/fisiologia , Estrogênios , Masculino , Área Pré-Óptica/metabolismo , Comportamento Sexual Animal/fisiologia , Testosterona/farmacologia
16.
Commun Biol ; 5(1): 383, 2022 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-35444217

RESUMO

Estrogen receptor (ER) α is involved in several estrogen-modulated neural and peripheral functions. To determine its role in the expression of female and male reproductive behavior, a mouse line lacking the ERα in the nervous system was generated. Mutant females did not exhibit sexual behavior despite normal olfactory preference, and had a reduced number of progesterone receptor-immunoreactive neurons in the ventromedial hypothalamus. Mutant males displayed a moderately impaired sexual behavior and unaffected fertility, despite evidences of altered organization of sexually dimorphic populations in the preoptic area. In comparison, males deleted for both neural ERα and androgen receptor (AR) displayed greater sexual deficiencies. Thus, these data highlight a predominant role for neural ERα in females and a complementary role with the AR in males in the regulation of sexual behavior, and provide a solid background for future analyses of neuronal versus glial implication of these signaling pathways in both sexes.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Comportamento Sexual Animal , Animais , Receptor alfa de Estrogênio/genética , Feminino , Hipotálamo/metabolismo , Masculino , Camundongos , Neurônios/metabolismo , Área Pré-Óptica/metabolismo
17.
Methods Mol Biol ; 2418: 53-61, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35119659

RESUMO

The ability to silence the expression of gene products in a chemically, spatially, and temporally specific manner in the brains of animals has enabled key breakthroughs in the field of behavioral neuroscience. Using this technique, estrogen receptor alpha (ERα) has been specifically implicated in a multitude of behaviors in mice, including sexual, aggressive, locomotor, and maternal behaviors, in a variety of brain regions, including the medial preoptic area, ventromedial hypothalamus, and amygdala. In this chapter, we describe the techniques involved in the generation of the small hairpin RNAs (shRNAs) specifically designed to silence ERα, the construction of the adeno-associated viral (AAV) vector for delivery of the shRNA, the procedures to confirm the silencing of ERα (in vitro and in vivo) and in vivo delivery of the shRNAs to the brains of animals.


Assuntos
Receptor alfa de Estrogênio , Roedores , Animais , Encéfalo/metabolismo , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Camundongos , Área Pré-Óptica/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Roedores/genética
18.
Horm Behav ; 141: 105129, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35168026

RESUMO

Maternal experience can promote a long-lasting increase in maternal motivation. This maintenance of caregiving behaviors, rather than avoidant or agnostic responses towards young, is advantageous for the survival of subsequent offspring. We have previously reported that maternal motivation is associated with differential immediate early gene expression in central motivation circuits and aversion circuits. Here we ask how these circuits come to differentially respond to infant cues. We used Targeted Recombination in Active Populations (TRAP) to identify cells that respond to pups in maternally hesitant TRAP2;Ai14 virgin female mice. Following an initial 60 min exposure to foster pups, virgin TRAP2;Ai14 mice were injected with 4-hydroxytamoxifen to induce recombination in c-Fos expressing cells and subsequent permanent expression of a red fluorescent reporter. We then examined whether the same cells that encode pup cues are reactivated during maternal memory retrieval two weeks later using c-Fos immunohistochemistry. Whereas initial pup exposure induced c-Fos activation exclusively in the medial preoptic area (MPOA), following repeated experience, c-Fos expression was significantly higher than baseline in multiple regions of maternal and central aversion circuits (e.g., ventral bed nucleus of the stria terminalis, nucleus accumbens, basolateral amygdala, prefrontal cortex, medial amygdala, and ventromedial nucleus of the hypothalamus). Further, cells in many of these sites were significantly reactivated during maternal memory retrieval. These data suggest that cells across both maternal motivation and central aversion circuits are stably responsive to pups and thus may form the cellular representation of maternal memory.


Assuntos
Comportamento Materno , Área Pré-Óptica , Animais , Feminino , Humanos , Hipotálamo/metabolismo , Comportamento Materno/fisiologia , Camundongos , Núcleo Accumbens/metabolismo , Área Pré-Óptica/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo
19.
Neurotoxicology ; 89: 41-54, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35026373

RESUMO

Nicotine is a neuroteratogenic component of tobacco smoke, e-cigarettes, and other products and can exert sex-specific effects in the developing brain, likely mediated through sex hormones. Estradiol modulates expression of nicotinic acetylcholine receptors in rats, and plays critical roles in neurodevelopmental processes, including sexual differentiation of the brain. Here, we examined the effects of developmental nicotine exposure on the sexual differentiation of the preoptic area (POA), a brain region that normally displays robust structural sexual dimorphisms and controls adult mating behavior in rodents. Using a rat model of gestational exposure, developing pups were exposed to nicotine (2 mg/kg/day) via maternal osmotic minipump (subcutaneously, sc) throughout the critical window for brain sexual differentiation. At postnatal day (PND) 4, a subset of offspring was analyzed for epigenetic effects in the POA. At PND40, all offspring were gonadectomized, implanted with a testosterone-releasing capsule (sc), and assessed for male sexual behavior at PND60. Following sexual behavior assessment, the area of the sexually dimorphic nucleus of the POA (SDN-POA) was measured using immunofluorescent staining techniques. In adults, normal sex differences in male sexual behavior and in the SDN-POA area were eliminated in nicotine-treated animals. Using novel analytical approaches to evaluate overall masculinization of the adult POA, we identified significant masculinization of the nicotine-treated female POA. In neonates (PND4), nicotine exposure induced trending alterations in methylation-dependent masculinizing gene expression and DNA methylation levels at sexually-dimorphic differentially methylated regions, suggesting that developmental nicotine exposure is capable of triggering masculinization of the rat POA via epigenetic mechanisms.


Assuntos
Sistemas Eletrônicos de Liberação de Nicotina , Área Pré-Óptica , Animais , Feminino , Masculino , Nicotina/toxicidade , Área Pré-Óptica/metabolismo , Ratos , Caracteres Sexuais , Diferenciação Sexual , Testosterona
20.
Neuroendocrinology ; 112(7): 649-665, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34592740

RESUMO

INTRODUCTION: Menopausal hot flashes are related to hypothalamic preoptic area (POA) dysfunction. Thermosensitive transient receptor potential channels (ThermoTRPs) are involved in temperature sensing and regulation of thermosensitive neurons (TSNs) in the POA. Whether ThermoTRP-TSNs in the POA, particularly the non-noxious thermoreceptor, transient receptor potential melastatin 2 (TRPM2), are involved in the occurrence of hot flashes is still unclear. METHODS: Twenty wild-type and 50 Trpm2-Cre adult female mice were randomly divided into sham (SHAM) and ovariectomy (OVX) groups. In the POA, ERα, ERß, GPR30, TRPA1, TRPM8, TRPM2, and TRPV1 expression was detected by Western blot or/and quantitative real-time polymerase chain reaction and the number of TSNs expressing TRPM2 (TRPM2-TSNs) by immunofluorescence. Before and after TRPM2-TSN activation/inhibition, back (BST) and tail skin temperature (TST) and the proportion of glutamatergic and GABAergic neurons among TRPM2-TSNs were recorded. RESULTS: Compared with SHAM, the expression of ERα, ERß, TRPM2, and TRPM8 in the POA of the OVX group decreased, with a significantly larger change range for TRPM2 than TRPM8. In addition, the number of TRPM2-TSNs showing TRPA1, TRPM8, and TRPV1 expression in the OVX group decreased, and the proportion of glutamatergic and GABAergic neurons in TRPM2-TSNs decreased and increased, respectively. Meanwhile, BST and TST increased. After activating or inhibiting TRPM2-TSNs, the proportions of glutamatergic and GABAergic neurons in TRPM2-TSNs changed, along with the BST and TST. CONCLUSION: In menopause, the abnormal quantity and function of TRPM2-TSNs in the POA is key for the development of hot flashes, characterized by an imbalance in heat dissipation and production due to the corresponding imbalance in glutamatergic and GABAergic neurons.


Assuntos
Canais de Cátion TRPM , Animais , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Feminino , Fogachos/metabolismo , Menopausa , Camundongos , Neurônios/metabolismo , Área Pré-Óptica/metabolismo , Canais de Cátion TRPM/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA