Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 165
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 18(11): e0294113, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37971993

RESUMO

Oxytocin (OXT) neurons project to various brain regions and its receptor expression is widely distributed. Although it has been reported that OXT administration affects cognitive function, it is unclear how endogenous OXT plays roles in cognitive function. The present study examined the role of endogenous OXT in mice cognitive function. OXT neurons were specifically activated by OXT neuron-specific excitatory Designer Receptors Exclusively Activated by Designer Drug expression system and following administration of clozapine-N-oxide (CNO). Object recognition memory was assessed with the novel object recognition task (NORT). Moreover, we observed the expression of c-Fos via immunohistochemical staining to confirm neuronal activity. In NORT, the novel object exploration time percentage significantly increased in CNO-treated mice. CNO-treated mice showed a significant increase in the number of c-Fos-positive cells in the supramammillary nucleus (SuM). In addition, we found that the OXT-positive fibers from paraventricular hypothalamic nucleus (PVN) were identified in the SuM. Furthermore, mice injected locally with CNO into the SuM to activate OXTergic axons projecting from the PVN to the SuM showed significantly increased percentage time of novel object exploration. Taken together, we proposed that object recognition memory in mice could be modulated by OXT neurons in the PVN projecting to the SuM.


Assuntos
Hipotálamo , Ocitocina , Animais , Camundongos , Hipotálamo/metabolismo , Ocitocina/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptores de Ocitocina/metabolismo , Hipotálamo Posterior/metabolismo , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo
2.
Sci Rep ; 13(1): 1447, 2023 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-36702854

RESUMO

Embryonic ethanol exposure in zebrafish and rats, while stimulating hypothalamic hypocretin/orexin (Hcrt) neurons along with alcohol consumption and related behaviors, increases the chemokine receptor Cxcr4 that promotes neuronal migration and may mediate ethanol's effects on neuronal development. Here we performed a more detailed anatomical analysis in zebrafish of ethanol's effects on the Cxcl12a/Cxcr4b system throughout the entire brain as it relates to Hcrt neurons developing within the anterior hypothalamus (AH) where they are normally located. We found that ethanol increased these Hcrt neurons only in the anterior part of the AH and induced ectopic Hcrt neurons further anterior in the preoptic area, and these effects along with ethanol-induced behaviors were completely blocked by a Cxcr4 antagonist. Analysis of cxcl12a transcripts and internalized Cxcr4b receptors throughout the brain showed they both exhibited natural posterior-to-anterior concentration gradients, with levels lowest in the posterior AH and highest in the anterior telencephalon. While stimulating their density in all areas and maintaining these gradients, ethanol increased chemokine expression only in the more anterior and ectopic Hcrt neurons, effects blocked by the Cxcr4 antagonist. These findings demonstrate how increased chemokine expression acting along natural gradients mediates ethanol-induced anterior migration of ectopic Hcrt neurons and behavioral disturbances.


Assuntos
Etanol , Peixe-Zebra , Animais , Ratos , Orexinas/metabolismo , Peixe-Zebra/metabolismo , Etanol/toxicidade , Etanol/metabolismo , Hipotálamo Posterior/metabolismo , Quimiocinas/metabolismo , Neurônios/metabolismo
3.
Sci Rep ; 13(1): 489, 2023 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-36627362

RESUMO

Posterior hypothalamus (PH), an important part of the descending pain processing pathway, has been found to be activated in trigeminal autonomic cephalalgias. However, there are very few studies conducted and information regarding its implications in trigeminal neuropathic pain (TNP). Therefore, we aimed to ascertain whether optogenetic inhibition of PH could affect the outcomes of a chronic constriction injury in the infraorbital nerve (CCI-ION) rat model. Animals were divided into the TNP animal, sham, and naive-control groups. CCI-ION surgery was performed to mimic TNP symptoms, and the optogenetic or null virus was injected into the ipsilateral PH. In vivo single-unit extracellular recordings were obtained from both the ipsilateral ventrolateral periaqueductal gray (vlPAG) and contralateral ventral posteromedial (VPM) thalamus in stimulation "OFF" and "ON" conditions. Alterations in behavioral responses during the stimulation-OFF and stimulation-ON states were examined. We observed that optogenetic inhibition of the PH considerably improved behavioral responses in TNP animals. We found increased and decreased firing activity in the vlPAG and VPM thalamus, respectively, during optogenetic inhibition of the PH. Inhibiting PH attenuates trigeminal pain signal transmission by modulating the vlPAG and trigeminal nucleus caudalis, thereby providing evidence of the therapeutic potential of PH in TNP management.


Assuntos
Neuralgia , Neuralgia do Trigêmeo , Ratos , Animais , Ratos Sprague-Dawley , Optogenética , Neuralgia/terapia , Neuralgia/metabolismo , Hipotálamo Posterior/metabolismo , Hiperalgesia/metabolismo
4.
Am J Physiol Regul Integr Comp Physiol ; 321(6): R938-R950, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34704845

RESUMO

Systemic administration of dopamine (DA) receptor agonists leads to falls in body temperature. However, the central thermoregulatory pathways modulated by DA have not been fully elucidated. Here we identified a source and site of action contributing to DA's hypothermic action by inhibition of brown adipose tissue (BAT) thermogenesis. Nanoinjection of the type 2 and type 3 DA receptor (D2R/D3R) agonist, 7-hydroxy-N,N-di-n-propyl-2-aminotetralin (7-OH-DPAT), in the rostral raphe pallidus area (rRPa) inhibits the sympathetic activation of BAT evoked by cold exposure or by direct activation of N-methyl-d-aspartate (NMDA) receptors in the rRPa. Blockade of D2R/D3R in the rRPa with nanoinjection of SB-277011A increases BAT thermogenesis, consistent with a tonic release of DA in the rRPa contributing to inhibition of BAT thermogenesis. Accordingly, D2Rs are expressed in cold-activated and serotonergic neurons in the rRPa, and anatomical tracing studies revealed that neurons in the posterior hypothalamus (PH) are a source of dopaminergic input to the rRPa. Disinhibitory activation of PH neurons with nanoinjection of gabazine inhibits BAT thermogenesis, which is reduced by pretreatment of the rRPa with SB-277011A. In conclusion, the rRPa, the site of sympathetic premotor neurons for BAT, receives a tonically active, dopaminergic input from the PH that suppresses BAT thermogenesis.


Assuntos
Tecido Adiposo Marrom/inervação , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Hipotálamo Posterior/metabolismo , Inibição Neural , Núcleo Pálido da Rafe/metabolismo , Termogênese , Animais , Agonistas de Dopamina/administração & dosagem , Neurônios Dopaminérgicos/efeitos dos fármacos , Antagonistas GABAérgicos/administração & dosagem , Hipotálamo Posterior/efeitos dos fármacos , Injeções , Masculino , Vias Neurais/metabolismo , Núcleo Pálido da Rafe/efeitos dos fármacos , Ratos Sprague-Dawley , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D3/metabolismo , Receptores de GABA-A/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Termogênese/efeitos dos fármacos
5.
Elife ; 102021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34468312

RESUMO

Escape from threats has paramount importance for survival. However, it is unknown if a single circuit controls escape vigor from innate and conditioned threats. Cholecystokinin (cck)-expressing cells in the hypothalamic dorsal premammillary nucleus (PMd) are necessary for initiating escape from innate threats via a projection to the dorsolateral periaqueductal gray (dlPAG). We now show that in mice PMd-cck cells are activated during escape, but not other defensive behaviors. PMd-cck ensemble activity can also predict future escape. Furthermore, PMd inhibition decreases escape speed from both innate and conditioned threats. Inhibition of the PMd-cck projection to the dlPAG also decreased escape speed. Intriguingly, PMd-cck and dlPAG activity in mice showed higher mutual information during exposure to innate and conditioned threats. In parallel, human functional magnetic resonance imaging data show that a posterior hypothalamic-to-dlPAG pathway increased activity during exposure to aversive images, indicating that a similar pathway may possibly have a related role in humans. Our data identify the PMd-dlPAG circuit as a central node, controlling escape vigor elicited by both innate and conditioned threats.


Assuntos
Comportamento Animal , Condicionamento Psicológico , Reação de Fuga , Medo , Hipotálamo Posterior/fisiologia , Substância Cinzenta Periaquedutal/fisiologia , Adulto , Animais , Mapeamento Encefálico , Colecistocinina/genética , Colecistocinina/metabolismo , Feminino , Humanos , Hipotálamo Posterior/diagnóstico por imagem , Hipotálamo Posterior/metabolismo , Imageamento por Ressonância Magnética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Vias Neurais/fisiologia , Optogenética , Substância Cinzenta Periaquedutal/diagnóstico por imagem , Substância Cinzenta Periaquedutal/metabolismo , Estimulação Luminosa , Ratos Long-Evans , Fatores de Tempo , Gravação em Vídeo , Percepção Visual , Adulto Jovem
6.
Brain Res ; 1748: 147125, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32931819

RESUMO

There was a recent report suggesting that LIM homeobox 6 (Lhx6)+ GABA-releasing neurons of the ventral zona incerta (ZI) promote sleep. We demonstrated in the previous study that Lhx6+ ZI neurons are activated during paradoxical sleep (PS) hypersomnia which was induced by 48-hour PS deprivation, implying their roles in the control of PS like melanin-concentrating hormone (MCH) cells. Since the core portion of the lateral supramammillary nucleus (SUMl) is the major hypothalamic area activating the dentate gyrus as well as other limbic cortices during PS, we examined in the present study whether Lhx6+ ZI cells provide efferent projections to the SUMl, using the retrograde-tracing method. The majority of Lhx6+ neurons projecting to the SUMl occupied the ventral border (or ventral one-third) of the ventral ZI. Based on the quantitative analysis, the mean number of retrogradely-labeled Lhx6+ neurons was comparable to that of retrogradely-labeled MCH cells in the ZI. However, the total (i.e., single- plus double-labeled) number of Lhx6+ cells was approximately three times larger than that of MCH cells in the ZI. Thus, the proportion (about 7.8%) of retrogradely-labeled Lhx6+ neurons over the total Lhx6+ cells was approximately one-third of the percentage (about 20.9%) of retrogradely-labeled MCH neurons over the total MCH cells. On the other hand, a combination of retrogradely-labeled, Lhx6 and MCH cells occupied approximately 43.7% of the total retrogradely-labeled neurons in the ventral ZI. The present observations suggested that Lhx6+ neurons in the ventral ZI might play an important role in the regulation of PS, partly via the neural network involving the SUMl.


Assuntos
Hipotálamo Posterior/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Fatores de Transcrição/metabolismo , Zona Incerta/metabolismo , Animais , Vias Neurais/metabolismo , Ratos , Ratos Sprague-Dawley
7.
Brain Struct Funct ; 224(8): 2983-2999, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31485718

RESUMO

Guanylyl cyclase C (GUCY2C) is the afferent central receptor in the gut-brain endocrine axis regulated by the anorexigenic intestinal hormone uroguanylin. GUCY2C mRNA and protein are produced in the hypothalamus, a major center regulating appetite and metabolic homeostasis. Further, GUCY2C mRNA and protein are expressed in the ventral midbrain, a principal structure regulating hedonic reward from behaviors including eating. While GUCY2C is expressed in hypothalamus and midbrain, its precise neuroanatomical organization and relationship with circuits regulating satiety remain unknown. Here, we reveal that hypothalamic GUCY2C mRNA is confined to the ventral premammillary nucleus (PMV), while in midbrain it is produced by neurons in the ventral tegmental area (VTA) and substantia nigra (SN). GUCY2C in the PMV is produced by 46% of neurons expressing anorexigenic leptin receptors, while in the VTA/SN it is produced in most tyrosine hydroxylase-immunoreactive neurons. In contrast to mRNA, GUCY2C protein is widely distributed throughout the brain in canonical sites of PMV and VTA/SN axonal projections. Selective stereotaxic ablation of PMV or VTA/SN neurons eliminated GUCY2C only in their respective canonical projection sites. Conversely, specific anterograde tracer analyses of PMV or VTA/SN neurons confirmed distinct GUCY2C-immunoreactive axons projecting to those canonical locations. Together, these findings reveal two discrete neuronal circuits expressing GUCY2C originating in the PMV in the hypothalamus and in the VTA/SN in midbrain, which separately project to other sites throughout the brain. They suggest a structural basis for a role for the GUCY2C-uroguanylin gut-brain endocrine axis in regulating homeostatic and behavioral components contributing to satiety.


Assuntos
Hipotálamo Posterior/metabolismo , Receptores de Enterotoxina/análise , Substância Negra/metabolismo , Área Tegmentar Ventral/metabolismo , Animais , Axônios , Feminino , Hipotálamo Posterior/citologia , Masculino , Camundongos Endogâmicos C57BL , Vias Neurais/citologia , RNA Mensageiro/análise , Substância Negra/citologia , Área Tegmentar Ventral/citologia
8.
Brain Struct Funct ; 224(1): 99-110, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30284033

RESUMO

Neurons containing melanin-concentrating hormone (MCH) in the posterior lateral hypothalamus play an integral role in rapid eye movement sleep (REMs) regulation. As MCH neurons also contain a variety of other neuropeptides [e.g., cocaine- and amphetamine-regulated transcript (CART) and nesfatin-1] and neurotransmitters (e.g., glutamate), the specific neurotransmitter responsible for REMs regulation is not known. We hypothesized that glutamate, the primary fast-acting neurotransmitter in MCH neurons, is necessary for REMs regulation. To test this hypothesis, we deleted vesicular glutamate transporter (Vglut2; necessary for synaptic release of glutamate) specifically from MCH neurons by crossing MCH-Cre mice (expressing Cre recombinase in MCH neurons) with Vglut2flox/flox mice (expressing LoxP-modified alleles of Vglut2), and studied the amounts, architecture and diurnal variation of sleep-wake states during baseline conditions. We then activated the MCH neurons lacking glutamate neurotransmission using chemogenetic methods and tested whether these MCH neurons still promoted REMs. Our results indicate that glutamate in MCH neurons contributes to normal diurnal variability of REMs by regulating the levels of REMs during the dark period, but MCH neurons can promote REMs even in the absence of glutamate.


Assuntos
Ritmo Circadiano , Ácido Glutâmico/metabolismo , Hormônios Hipotalâmicos/metabolismo , Hipotálamo Posterior/metabolismo , Melaninas/metabolismo , Neurônios/metabolismo , Hormônios Hipofisários/metabolismo , Sono REM , Animais , Hormônios Hipotalâmicos/genética , Hipotálamo Posterior/citologia , Masculino , Melaninas/genética , Camundongos Transgênicos , Fotoperíodo , Hormônios Hipofisários/genética , Fatores de Tempo , Proteína Vesicular 2 de Transporte de Glutamato/genética , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo , Vigília
9.
J Neuroendocrinol ; 30(1)2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29224246

RESUMO

Among vertebrate species of the major vertebrate classes in the wild, a seasonal rhythm of whole body fuel metabolism, oscillating from a lean to obese condition, is a common biological phenomenon. This annual cycle is driven in part by annual changes in the circadian dopaminergic signalling at the suprachiasmatic nuclei (SCN), with diminution of circadian peak dopaminergic activity at the SCN facilitating development of the seasonal obese insulin-resistant condition. The present study investigated whether such an ancient circadian dopamine-SCN activity system for expression of the seasonal obese, insulin-resistant phenotype may be operative in animals made obese amd insulin resistant by high-fat feeding and, if so, whether reinstatement of the circadian dopaminergic peak at the SCN would be sufficient to reverse the adverse metabolic impact of the high-fat diet without any alteration of caloric intake. First, we identified the supramammillary nucleus as a novel site providing the majority of dopaminergic neuronal input to the SCN. We further identified dopamine D2 receptors within the peri-SCN region as being functional in mediating SCN responsiveness to local dopamine. In lean, insulin-sensitive rats, the peak in the circadian rhythm of dopamine release at the peri-SCN coincided with the daily peak in SCN electrophysiological responsiveness to local dopamine administration. However, in rats made obese and insulin resistant by high-fat diet (HFD) feeding, these coincident circadian peak activities were both markedly attenuated or abolished. Reinstatement of the circadian peak in dopamine level at the peri-SCN by its appropriate circadian-timed daily microinjection to this area (but not outside this circadian time-interval) abrogated the obese, insulin-resistant condition without altering the consumption of the HFD. These findings suggest that the circadian peak of dopaminergic activity at the peri-SCN/SCN is a key modulator of metabolism and the responsiveness to adverse metabolic consequences of HFD consumption.


Assuntos
Ritmo Circadiano/fisiologia , Dieta Hiperlipídica , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Núcleo Supraquiasmático/metabolismo , Animais , Relógios Biológicos , Feminino , Hipotálamo Posterior/metabolismo , Obesidade/metabolismo , Ratos , Ratos Endogâmicos SHR , Ratos Sprague-Dawley , Receptores de Dopamina D2/metabolismo
10.
Eur Neuropsychopharmacol ; 27(11): 1120-1131, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28939165

RESUMO

Acute γ-aminobutyric acid (GABA) disinhibition in the posterior hypothalamus (PH) elicits defensive reactions that are considered anxiety- and panic attack-like behaviour, and these defensive reactions are followed by antinociception. Evidence indicates that the PH connects with the medial prefrontal cortex, particularly the anterior cingulate cortex (ACC), which seems to regulate these unconditioned fear-induced defensive responses. However, few studies have shown the participation of cortical regions in the control of behavioural and antinociceptive responses organised by diencephalic structures. It has been suggested that the glutamatergic system can mediate this cortical influence, as excitatory imbalance is believed to play a role in both defensive mechanisms. Thus, the aim of the present study was to investigate the involvement of ACC glutamatergic connections via blockade of local N-methyl-D-aspartate (NMDA) receptors to elaborate panic-like defensive behaviours and unconditioned fear-induced antinociception organised by PH neurons. Wistar rats were treated with microinjections of 0.9% NaCl or LY235959 (a selective NMDA receptor antagonist) in the ACC at different concentrations (2, 4 and 8 nmol/0.2µL), followed by GABAA receptor blockade in the PH. Defensive reactions were analysed for 20min, and the nociceptive threshold was then measured at 10-min intervals for 60min. Pretreatment of the ACC with LY235959 reduced both panic-like defensive behaviour and fear-induced antinociception evoked by PH GABAergic disinhibition. Our findings suggest that ACC NMDA receptor-signalled glutamatergic inputs play a relevant role in the organisation of anxiety- and panic attack-like behaviours and in fear-induced antinociception.


Assuntos
Reação de Fuga/fisiologia , Medo/fisiologia , Giro do Cíngulo/metabolismo , Hipotálamo Posterior/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais/fisiologia , Análise de Variância , Animais , Bicuculina/análogos & derivados , Bicuculina/farmacologia , Relação Dose-Resposta a Droga , Reação de Fuga/efeitos dos fármacos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Medo/efeitos dos fármacos , Antagonistas de Receptores de GABA-A/farmacologia , Giro do Cíngulo/efeitos dos fármacos , Hipotálamo Posterior/efeitos dos fármacos , Isoquinolinas/farmacologia , Masculino , Microinjeções , Medição da Dor , Pânico/efeitos dos fármacos , Pânico/fisiologia , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos
11.
Endocr J ; 64(1): 7-14, 2017 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-27665725

RESUMO

Hypersecretion of luteinizing hormone (LH) is a common endocrinological finding of polycystic ovary syndrome (PCOS). This derangement might have a close relationship with hypothalamic kisspeptin expression that is thought to be a key regulator of gonadotropin-releasing hormone (GnRH). We evaluated the relationship between the hypothalamic-pituitary-gonadal axis (HPG axis) and kisspeptin using a rat model of PCOS induced by letrozole. Letrozole pellets (0.4 mg/day) and control pellets were placed subcutaneously onto the backs of 3-week-old female Wistar rats. Body weight, vaginal opening and vaginal smear were checked daily. Blood and tissues of ovary, uterus and brain were collected at 12-weeks of age. An hypothalamic block was cut into anterior and posterior blocks, which included the anteroventral periventricular nucleus (AVPV) and the arcuate nucleus (ARC), respectively, in order to estimate hypothalamic kisspeptin expression in each area. The letrozole group showed a similar phenotype to human PCOS such as heavier body weight, heavier ovary, persistent anovulatory state, multiple enlarged follicles with no corpus luteum and higher LH and testosterone (T) levels compared to the control group. Kisspeptin mRNA expression in the posterior hypothalamic block including ARC was higher in the letrozole group than in the control group although its expression in the anterior hypothalamic block was similar between groups. These results suggest that enhanced KNDy neuron activity in ARC contributes to hypersecretion of LH in PCOS and might be a therapeutic target to rescue ovulatory disorder of PCOS in the future.


Assuntos
Hipotálamo Posterior/metabolismo , Kisspeptinas/genética , Síndrome do Ovário Policístico/genética , Animais , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Humanos , Hipotálamo Posterior/patologia , Kisspeptinas/metabolismo , Letrozol , Nitrilas , Síndrome do Ovário Policístico/induzido quimicamente , Síndrome do Ovário Policístico/patologia , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Triazóis , Regulação para Cima
12.
Sci Rep ; 6: 32877, 2016 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-27599613

RESUMO

Nesfatin-1 is an important molecule in the regulation of reproduction. However, its role in the reproductive axis in male animals remains to be understood. Here, we found that nesfatin-1 was mainly distributed in the arcuate nucleus (ARC), paraventricular nucleus (PVN), periventricular nucleus (PeN), and lateral hypothalamic area (LHA) of the hypothalamus; adenohypophysis and Leydig cells in male rats. Moreover, the concentrations of serum nesfatin-1 and its mRNA in hypothalamo-pituitary-gonadal axis (HPGA) vary with the age of the male rat. After intracerebroventricular injection of nesfatin-1, the hypothalamic genes for gonadotrophin releasing hormone (GnRH), kisspeptin (Kiss-1), pituitary genes for follicle-stimulate hormone ß(FSHß), luteinizing hormone ß(LHß), and genes for testicular steroidogenic acute regulatory (StAR) expression levels were decreased significantly. Nesfatin-1 significantly increased the expression of genes for 3ß-hydroxysteroid dehydrogenase (3ß-HSD), 17ß-hydroxysteroid dehydrogenase (17ß-HSD), and cytochrome P450 cleavage (P450scc) in the testis of pubertal rats, but their levels decreased in adult rats (P < 0.05), along with the serum FSH, LH, and testosterone (T) concentrations. After nesfatin-1 addition in vitro, T concentrations of the supernatant were significantly higher than that in the control group. These results were suggestive of the role of nesfatin-1 in the regulation of the reproductive axis in male rats.


Assuntos
Proteínas de Ligação ao Cálcio/fisiologia , Proteínas de Ligação a DNA/fisiologia , Hipotálamo/metabolismo , Células Intersticiais do Testículo/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Adeno-Hipófise/metabolismo , 17-Hidroxiesteroide Desidrogenases/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Hormônio Foliculoestimulante/metabolismo , Subunidade beta do Hormônio Folículoestimulante/metabolismo , Hipotálamo Posterior/metabolismo , Hormônio Luteinizante/metabolismo , Masculino , Nucleobindinas , Núcleo Hipotalâmico Paraventricular/metabolismo , Hipófise/metabolismo , Ratos , Testículo/metabolismo , Testosterona/metabolismo
13.
Neuropharmacology ; 110(Pt A): 396-406, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27496691

RESUMO

The obesity epidemic continues unabated and currently available pharmacological treatments are not sufficiently effective. Combining gut/brain peptide, GLP-1, with estrogen into a conjugate may represent a novel, safe and potent, strategy to treat diabesity. Here we demonstrate that the central administration of GLP-1-estrogen conjugate reduced food reward, food intake, and body weight in rats. In order to determine the brain location of the interaction of GLP-1 with estrogen, we avail of single-photon emission computed tomography imaging of regional cerebral blood flow and pinpoint a brain site unexplored for its role in feeding and reward, the supramammillary nucleus (SUM) as a potential target of the conjugated GLP-1-estrogen. We confirm that conjugated GLP-1 and estrogen directly target the SUM with site-specific microinjections. Additional microinjections of GLP-1-estrogen into classic energy balance controlling nuclei, the lateral hypothalamus (LH) and the nucleus of the solitary tract (NTS) revealed that the metabolic benefits resulting from GLP-1-estrogen injections are mediated through the LH and to some extent by the NTS. In contrast, no additional benefit of the conjugate was noted on food reward when the compound was microinjected into the LH or the NTS, identifying the SUM as the only neural substrate identified here to underlie the reward reducing benefits of GLP-1 and estrogen conjugate. Collectively we discover a surprising neural substrate underlying food intake and reward effects of GLP-1 and estrogen and uncover a new brain area capable of regulating energy balance and reward.


Assuntos
Peso Corporal/fisiologia , Estrogênios/metabolismo , Alimentos , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Hipotálamo Posterior/metabolismo , Recompensa , Animais , Mapeamento Encefálico , Fármacos do Sistema Nervoso Central/farmacologia , Circulação Cerebrovascular/fisiologia , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Estrogênios/administração & dosagem , Peptídeo 1 Semelhante ao Glucagon/administração & dosagem , Hipotálamo Posterior/diagnóstico por imagem , Hipotálamo Posterior/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Motivação/efeitos dos fármacos , Motivação/fisiologia , Ratos Sprague-Dawley , Tomografia Computadorizada de Emissão de Fóton Único
14.
Peptides ; 81: 9-14, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27083313

RESUMO

Hypocretin-1 and 2 (HCRT-1 and HCRT-2, respectively) are neuropeptides synthesized by neurons located in the postero-lateral hypothalamus, whose projections are widely distributed throughout the brain. The hypocretinergic (HCRTergic) system has been associated with the generation and maintenance of wakefulness, as well as with the promotion of motivated behaviors. In lactating rats, intra-cerebroventricular HCRT-1 administration stimulates maternal behavior, whilst lactation per se increases the expression of HCRT type 1 receptor (HCRT-R1). Due to the fact that HCRTergic receptors are expressed in the medial preoptic area (mPOA), a region critically involved in maternal behavior, we hypothesize that HCRT-1 promotes maternal behavior acting on this region. In order to evaluate this hypothesis, we assessed the maternal behavior of lactating rats following microinjections of HCRT-1 (10 or 100µM) and the selective HCRT-R1 antagonist SB-334867 (250µM) into the mPOA, during the first and second postpartum weeks. While intra-mPOA microinjections of HCRT-1 (100µM) increased corporal pup licking during the second postpartum week, the blockade of HCRT-R1 significantly decreased active components of maternal behavior, such as retrievals, corporal and ano-genital lickings, and increased the time spent in nursing postures in both postpartum periods. We conclude that HCRTergic system in the mPOA may stimulate maternal behavior, suggesting that endogenous HCRT-1 is necessary for the natural display of this behavior.


Assuntos
Lactação/metabolismo , Comportamento Materno/fisiologia , Receptores de Orexina/fisiologia , Orexinas/fisiologia , Área Pré-Óptica/metabolismo , Animais , Escala de Avaliação Comportamental , Benzoxazóis/farmacologia , Feminino , Região Hipotalâmica Lateral/citologia , Região Hipotalâmica Lateral/metabolismo , Hipotálamo Posterior/citologia , Hipotálamo Posterior/metabolismo , Infusões Intraventriculares , Comportamento Materno/efeitos dos fármacos , Microinjeções , Naftiridinas , Neurônios/metabolismo , Receptores de Orexina/metabolismo , Orexinas/administração & dosagem , Orexinas/farmacologia , Área Pré-Óptica/efeitos dos fármacos , Ratos , Ratos Wistar , Estatísticas não Paramétricas , Ureia/análogos & derivados , Ureia/farmacologia
15.
Gen Comp Endocrinol ; 230-231: 57-66, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-27055929

RESUMO

The premammillary nucleus (PMM) of the turkey mediobasal hypothalamus, where dopamine-melatonin (DA-Mel) neurons are localized, is a site for photoreception and photoperiodic time measurement, which is essential for the initiation of avian reproductive seasonality. In addition, this area could also be responsible for the onset and maintenance of photorefractoriness at the end of the breeding season due to the enhanced inhibitory effect of γ-aminobutyric acid (GABA). GABA is an inhibitory neurotransmitter in the central nervous system which interferes with the photosexual response in the turkey, a seasonally breeding bird. Here, we further characterized the GABAA receptor subunits in the PMM DA-Mel neurons related to reproductive seasonality and the onset of photorefractoriness. GABAA receptor subunits and GABA synthesis enzymes in the PMM of photosensitive and photorefractory turkey hens were identified using real-time qRT-PCR. The upregulation of GABAA receptor α1-3, ß2-3, γ1-3, ρ1-3, δ, and θ mRNA expression were observed in the PMM of photorefractory birds when compared to those of photosensitive ones while there is no change observed in the GABA synthesis enzymes, glutamate decarboxylase 1 and 2. Those upregulated GABAA receptor subunits were further examined using immunohistochemical staining and they appeared to be co-localized within the PMM DA-Mel neurons. The upregulation of GABAA receptor subunits observed in the PMM of photorefractory birds coincides with a lack of responsiveness to a light stimulus provided during the photosensitive phase. This is supported by the absence of c-fos induction and TH upregulation in the PMM and a subsequence inhibition of c-fos and GnRH-I expression in the nucleus commissurae pallii. The augmented GABAA receptor subunits expression may mediate an enhancement of inhibitory GABAergic neurotransmission and the subsequent interference with the photosexual response. This could contribute to the state of photorefractoriness and the termination of breeding activities in the turkey, a temperate zone bird.


Assuntos
Neurônios GABAérgicos/metabolismo , Hipotálamo Posterior/metabolismo , Hipotálamo Posterior/efeitos da radiação , Luz , Receptores de GABA-A/metabolismo , Perus/metabolismo , Regulação para Cima/efeitos da radiação , Animais , Arilalquilamina N-Acetiltransferase/metabolismo , Dopamina/metabolismo , Feminino , Hormônio Liberador de Gonadotropina/genética , Melatonina/metabolismo , Fotoperíodo , Precursores de Proteínas/genética , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de GABA-A/química , Reprodução/fisiologia , Reprodução/efeitos da radiação , Estações do Ano , Transmissão Sináptica , Triptofano Hidroxilase/metabolismo , Ácido gama-Aminobutírico/metabolismo
16.
Sci Rep ; 6: 20206, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26831220

RESUMO

Corticotropin releasing factor (CRF), a peptide hormone involved in the stress response, holds a key position in cardiovascular regulation. Here, we report that the central effect of CRF on cardiovascular activities is mediated by the posterior hypothalamic nucleus (PH), an important structure responsible for stress-induced cardiovascular changes. Our present results demonstrate that CRF directly excites PH neurons via two CRF receptors, CRFR1 and CRFR2, and consequently increases heart rate (HR) rather than the mean arterial pressure (MAP) and renal sympathetic nerve activity (RSNA). Bilateral vagotomy does not influence the tachycardia response to microinjection of CRF into the PH, while ß adrenergic receptor antagonist propranolol almost totally abolishes the tachycardia. Furthermore, microinjecting CRF into the PH primarily increases neuronal activity of the rostral ventrolateral medulla (RVLM) and rostral ventromedial medulla (RVMM), but does not influence that of the dorsal motor nucleus of the vagus nerve (DMNV). These findings suggest that the PH is a critical target for central CRF system in regulation of cardiac activity and the PH-RVLM/RVMM-cardiac sympathetic nerve pathways, rather than PH-DMNV-vagus pathway, may contribute to the CRF-induced tachycardia.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Hipotálamo Posterior/citologia , Hipotálamo Posterior/metabolismo , Neurônios/metabolismo , Taquicardia/etiologia , Taquicardia/metabolismo , Animais , Pressão Sanguínea , Hormônio Liberador da Corticotropina/farmacologia , Expressão Gênica , Frequência Cardíaca , Hipotálamo Posterior/efeitos dos fármacos , Rim/efeitos dos fármacos , Rim/inervação , Masculino , Bulbo/efeitos dos fármacos , Bulbo/metabolismo , Microinjeções , Neurônios/efeitos dos fármacos , Ratos , Receptores de Hormônio Liberador da Corticotropina/genética , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Sistema Nervoso Simpático , Taquicardia/fisiopatologia , Vagotomia
17.
J Neurosci ; 36(3): 795-805, 2016 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-26791210

RESUMO

A likely adaptive process mitigating the effects of chronic stress is the phenomenon of stress habituation, which frequently reduces multiple stress-evoked responses to the same (homotypic) stressor experienced repeatedly. The current studies investigated putative brain circuits that may coordinate the reduction of stress-related responses associated with stress habituation, a process that is inadequately understood. Initially, two rat premotor regions that respectively regulate neuroendocrine (medial parvicellular region of the paraventricular hypothalamic nucleus [PaMP]) and autonomic (rostral medullary raphe pallidus [RPa]) responses were targeted with distinguishable retrograde tracers. Two to 3 weeks later, injected animals underwent loud noise stress, and their brains were processed for fluorescent immunohistochemical detection of the tracers and the immediate early gene Fos. A rostral region of the posterior hypothalamic nucleus (rPH), and to a lesser extent, the median preoptic nucleus, exhibited the highest numbers of retrogradely labeled cells from both the RPa and PaMP that were colocalized with loud noise-induced Fos expression. Injections of an anterograde tracer in the rPH confirmed these connections and suggested that this region may contribute to the coordination of multiple stress-related responses. This hypothesis was partially tested by posterior hypothalamic injections of small volumes of muscimol, which disrupts normal synaptic functions, before acute and repeated loud noise or restraint exposures. In addition to significantly reduced corticosterone release in response to these two distinct stressors, rPH muscimol disrupted habituation to each stressor modality, suggesting a novel and important contribution of the rostral posterior hypothalamic nucleus in this category of adaptive processes. Significance statement: Habituation to stress is a process that possibly diminishes the detrimental health consequences of chronic stress by reducing the amplitude of many responses when the same challenging conditions are experienced repeatedly. Stress elicits a highly coordinated set of neuroendocrine, autonomic, and behavioral responses that are independently and relatively well defined; however, how the brain achieves coordination of these responses and their habituation-related declines is not well understood. The current studies provide some of the first anatomical and functional results suggesting that a specific region of the hypothalamus, the rostral posterior hypothalamic nucleus, targets multiple premotor regions and contributes to the regulation of acute neuroendocrine responses and their habituation to repeated stress.


Assuntos
Sistema Hipotálamo-Hipofisário/fisiologia , Hipotálamo Posterior/metabolismo , Sistema Hipófise-Suprarrenal/fisiologia , Estresse Psicológico/metabolismo , Doença Aguda , Animais , Sistema Hipotálamo-Hipofisário/química , Hipotálamo Posterior/química , Masculino , Sistema Hipófise-Suprarrenal/química , Ratos , Ratos Sprague-Dawley , Estresse Psicológico/psicologia
18.
Endocrinology ; 156(7): 2595-607, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25860032

RESUMO

Pheromones induce sexually dimorphic neuroendocrine responses, such as LH secretion. However, the neuronal network by which pheromones are converted into signals that will initiate and modulate endocrine changes remains unclear. We asked whether 2 sexually dimorphic populations in the anteroventral periventricular and periventricular nuclei that express kisspeptin and tyrosine hydroxylase (TH) are potential candidates that will transduce the olfactory signal to the neuroendocrine system. Furthermore, we assessed whether this transduction is sensitive to perinatal actions of estradiol by using female mice deficient in α-fetoprotein (AfpKO), which lack the protective actions of Afp against maternal estradiol. Wild-type (WT) and AfpKO male and female mice were exposed to same- versus opposite-sex odors and the expression of Fos (the protein product of the immediate early gene c-Fos) was analyzed along the olfactory projection pathways as well as whether kisspeptin, TH, and GnRH neurons are responsive to opposite-sex odors. Male odors induced a female-typical Fos expression in target forebrain sites of olfactory inputs involved in reproduction in WT, but not in AfpKO females, whereas female odors induced a male-typical Fos expression in males of both genotypes. In WT females, opposite-sex odors induced Fos in kisspeptin and TH neurons, whereas in AfpKO females and WT males, only a lower, but still significant, Fos expression was observed in TH but not in kisspeptin neurons. Finally, opposite-sex odors did not induce any significant Fos expression in GnRH neurons of both sexes or genotypes. Our results strongly suggest a role for fetal estrogen in the sexual differentiation of neural responses to sex-related olfactory cues.


Assuntos
Estradiol/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Neurônios/metabolismo , Atrativos Sexuais/fisiologia , Comportamento Sexual Animal/fisiologia , Animais , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/citologia , Hipotálamo Anterior/citologia , Hipotálamo Anterior/metabolismo , Hipotálamo Posterior/citologia , Hipotálamo Posterior/metabolismo , Camundongos , Camundongos Knockout , Neurônios/citologia , Odorantes , Prosencéfalo/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Transdução de Sinais/genética , Tirosina 3-Mono-Oxigenase/metabolismo , alfa-Fetoproteínas/genética
19.
Analyst ; 140(11): 3759-65, 2015 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-25902865

RESUMO

Histamine plays a major role in the mediation of allergic reactions such as peripheral inflammation. This classical monoamine is also a neurotransmitter involved in the central nervous system but its role in this context is poorly understood. Studying histamine neurotransmission is important due to its implications in many neurological disorders. The sensitivity, selectivity and high temporal resolution of fast scan cyclic voltammetry (FSCV) offer many advantages for studying electroactive neurotransmitters. Histamine has previously been studied with FSCV; however, the lack of a robust Faradaic electrochemical signal makes it difficult to selectively identify histamine in complex media, as found in vivo. In this work, we optimize an electrochemical waveform that provides a stimulation-locked and unique electrochemical signal towards histamine. We describe in vitro waveform optimization and a novel in vivo physiological model for stimulating histamine release in the mouse premammillary nucleus via stimulation of the medial forebrain bundle. We demonstrate that a robust signal can be used to effectively identify histamine and characterize its in vivo kinetics.


Assuntos
Eletroquímica/métodos , Liberação de Histamina , Histamina/química , Hipotálamo Posterior/metabolismo , Adsorção , Animais , Carbono/química , Fibra de Carbono , Condutividade Elétrica , Estimulação Elétrica , Eletroquímica/instrumentação , Masculino , Feixe Prosencefálico Mediano , Camundongos , Microeletrodos
20.
Endocrinology ; 156(7): 2582-94, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25856430

RESUMO

Kisspeptin neurons play an essential role in the regulation of fertility through direct regulation of the GnRH neurons. However, the relative contributions of the two functionally distinct kisspeptin neuron subpopulations to this critical regulation are not fully understood. Here we analyzed the specific projection patterns of kisspeptin neurons originating from either the rostral periventricular nucleus of the third ventricle (RP3V) or the arcuate nucleus (ARN) using a cell-specific, viral-mediated tract-tracing approach. We stereotaxically injected a Cre-dependent recombinant adenovirus encoding farnesylated enhanced green fluorescent protein into the ARN or RP3V of adult male and female mice expressing Cre recombinase in kisspeptin neurons. Fibers from ARN kisspeptin neurons projected widely; however, we did not find any evidence for direct contact with GnRH neuron somata or proximal dendrites in either sex. In contrast, we identified RP3V kisspeptin fibers in close contact with GnRH neuron somata and dendrites in both sexes. Fibers originating from both the RP3V and ARN were observed in close contact with distal GnRH neuron processes in the ARN and in the lateral and internal aspects of the median eminence. Furthermore, GnRH nerve terminals were found in close contact with the proximal dendrites of ARN kisspeptin neurons in the ARN, and ARN kisspeptin fibers were found contacting RP3V kisspeptin neurons in both sexes. Together these data delineate selective zones of kisspeptin neuron inputs to GnRH neurons and demonstrate complex interconnections between the distinct kisspeptin populations and GnRH neurons.


Assuntos
Dendritos/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/citologia , Kisspeptinas/metabolismo , Neurônios/citologia , Adenoviridae , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/metabolismo , Feminino , Proteínas de Fluorescência Verde , Hipotálamo/metabolismo , Hipotálamo Anterior/citologia , Hipotálamo Anterior/metabolismo , Hipotálamo Posterior/citologia , Hipotálamo Posterior/metabolismo , Masculino , Camundongos , Neurônios/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...