Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.870
Filtrar
1.
Genes Brain Behav ; 23(3): e12906, 2024 06.
Artigo em Inglês | MEDLINE | ID: mdl-38861664

RESUMO

Motherhood is a costly life-history transition accompanied by behavioral and neural plasticity necessary for offspring care. Motherhood in the monogamous prairie vole is associated with decreased pair bond strength, suggesting a trade-off between parental investment and pair bond maintenance. Neural mechanisms governing pair bonds and maternal bonds overlap, creating possible competition between the two. We measured mRNA expression of genes encoding receptors for oxytocin (oxtr), dopamine (d1r and d2r), mu-opioids (oprm1a), and kappa-opioids (oprk1a) within three brain areas processing salience of sociosensory cues (anterior cingulate cortex; ACC), pair bonding (nucleus accumbens; NAc), and maternal care (medial preoptic area; MPOA). We compared gene expression differences between pair bonded prairie voles that were never pregnant, pregnant (~day 16 of pregnancy), and recent mothers (day 3 of lactation). We found greater gene expression in the NAc (oxtr, d2r, oprm1a, and oprk1a) and MPOA (oxtr, d1r, d2r, oprm1a, and oprk1a) following the transition to motherhood. Expression for all five genes in the ACC was greatest for females that had been bonded for longer. Gene expression within each region was highly correlated, indicating that oxytocin, dopamine, and opioids comprise a complimentary gene network for social signaling. ACC-NAc gene expression correlations indicated that being a mother (oxtr and d1r) or maintaining long-term pair bonds (oprm1a) relies on the coordination of different signaling systems within the same circuit. Our study suggests the maternal brain undergoes changes that prepare females to face the trade-off associated with increased emotional investment in offspring, while also maintaining a pair bond.


Assuntos
Arvicolinae , Comportamento Materno , Núcleo Accumbens , Ligação do Par , Receptores Opioides mu , Animais , Feminino , Arvicolinae/genética , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Comportamento Materno/fisiologia , Núcleo Accumbens/metabolismo , Gravidez , Receptores de Ocitocina/genética , Receptores de Ocitocina/metabolismo , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Giro do Cíngulo/metabolismo , Área Pré-Óptica/metabolismo , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo
2.
Cells ; 13(11)2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38891027

RESUMO

Sleep disruption is a frequent problem of advancing age, often accompanied by low-grade chronic central and peripheral inflammation. We examined whether chronic neuroinflammation in the preoptic and basal forebrain area (POA-BF), a critical sleep-wake regulatory structure, contributes to this disruption. We developed a targeted viral vector designed to overexpress tumor necrosis factor-alpha (TNFα), specifically in astrocytes (AAV5-GFAP-TNFα-mCherry), and injected it into the POA of young mice to induce heightened neuroinflammation within the POA-BF. Compared to the control (treated with AAV5-GFAP-mCherry), mice with astrocytic TNFα overproduction within the POA-BF exhibited signs of increased microglia activation, indicating a heightened local inflammatory milieu. These mice also exhibited aging-like changes in sleep-wake organization and physical performance, including (a) impaired sleep-wake functions characterized by disruptions in sleep and waking during light and dark phases, respectively, and a reduced ability to compensate for sleep loss; (b) dysfunctional VLPO sleep-active neurons, indicated by fewer neurons expressing c-fos after suvorexant-induced sleep; and (c) compromised physical performance as demonstrated by a decline in grip strength. These findings suggest that inflammation-induced dysfunction of sleep- and wake-regulatory mechanisms within the POA-BF may be a critical component of sleep-wake disturbances in aging.


Assuntos
Envelhecimento , Astrócitos , Prosencéfalo Basal , Área Pré-Óptica , Sono , Fator de Necrose Tumoral alfa , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Envelhecimento/metabolismo , Área Pré-Óptica/metabolismo , Camundongos , Fator de Necrose Tumoral alfa/metabolismo , Sono/fisiologia , Prosencéfalo Basal/metabolismo , Prosencéfalo Basal/patologia , Vigília , Masculino , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Neurônios/patologia , Transtornos do Sono-Vigília/metabolismo , Transtornos do Sono-Vigília/patologia
3.
J Physiol Sci ; 74(1): 33, 2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38867187

RESUMO

Hibernation and torpor are not passive responses caused by external temperature drops and fasting but are active brain functions that lower body temperature. A population of neurons in the preoptic area was recently identified as such active torpor-regulating neurons. We hypothesized that the other hypothermia-inducing maneuvers would also activate these neurons. To test our hypothesis, we first refined the previous observations, examined the brain regions explicitly activated during the falling phase of body temperature using c-Fos expression, and confirmed the preoptic area. Next, we observed long-lasting hypothermia by reactivating torpor-tagged Gq-expressing neurons using the activity tagging and DREADD systems. Finally, we found that about 40-60% of torpor-tagged neurons were activated by succeeding isoflurane anesthesia and by icv administration of an adenosine A1 agonist. Isoflurane-induced and central adenosine-induced hypothermia is, at least in part, an active process mediated by the torpor-regulating neurons in the preoptic area.


Assuntos
Adenosina , Isoflurano , Neurônios , Área Pré-Óptica , Animais , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/metabolismo , Isoflurano/farmacologia , Isoflurano/administração & dosagem , Adenosina/administração & dosagem , Adenosina/farmacologia , Adenosina/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/fisiologia , Masculino , Anestésicos Inalatórios/farmacologia , Anestésicos Inalatórios/administração & dosagem , Temperatura Corporal/efeitos dos fármacos , Temperatura Corporal/fisiologia , Hipotermia/induzido quimicamente , Hipotermia/metabolismo , Torpor/efeitos dos fármacos , Camundongos , Proteínas Proto-Oncogênicas c-fos/metabolismo
4.
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
5.
Cell Rep ; 43(5): 114192, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38703367

RESUMO

The preoptic area of the hypothalamus (POA) is essential for sleep regulation. However, the cellular makeup of the POA is heterogeneous, and the molecular identities of the sleep-promoting cells remain elusive. To address this question, this study compares mice during recovery sleep following sleep deprivation to mice allowed extended sleep. Single-nucleus RNA sequencing (single-nucleus RNA-seq) identifies one galanin inhibitory neuronal subtype that shows upregulation of rapid and delayed activity-regulated genes during recovery sleep. This cell type expresses higher levels of growth hormone receptor and lower levels of estrogen receptor compared to other galanin subtypes. single-nucleus RNA-seq also reveals cell-type-specific upregulation of purinergic receptor (P2ry14) and serotonin receptor (Htr2a) during recovery sleep in this neuronal subtype, suggesting possible mechanisms for sleep regulation. Studies with RNAscope validate the single-nucleus RNA-seq findings. Thus, the combined use of single-nucleus RNA-seq and activity-regulated genes identifies a neuronal subtype functionally involved in sleep regulation.


Assuntos
Galanina , Neurônios , Área Pré-Óptica , Privação do Sono , Animais , Galanina/metabolismo , Galanina/genética , Neurônios/metabolismo , Área Pré-Óptica/metabolismo , Camundongos , Privação do Sono/metabolismo , Privação do Sono/genética , Masculino , RNA-Seq , Camundongos Endogâmicos C57BL , Sono/genética , Sono/fisiologia , Análise de Célula Única
6.
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
7.
Physiol Rep ; 12(10): e16046, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38749925

RESUMO

We have previously reported that the subfornical organ (SFO) does not contribute to the chronic hypertensive response to DOCA-salt in rats, and yet the organum vasculosum of the lamina terminalis (OVLT) plays a significant role in the development of deoxycorticosterone acetate (DOCA)-salt hypertension. Since efferent fibers of the OVLT project to and through the median preoptic nucleus (MnPO), the present study was designed to test the hypothesis that the MnPO is necessary for DOCA-salt hypertension in the rat. Male Sprague-Dawley rats underwent SHAM (MnPOsham; n = 5) or electrolytic lesion of the MnPO (MnPOx; n = 7) followed by subsequent unilateral nephrectomy and telemetry instrumentation. After recovery and during the experimental protocol, rats consumed a 0.1% NaCl diet and 0.9% NaCl drinking solution. Mean arterial pressure (MAP) was recorded telemetrically 5 days before and 21 days after DOCA implantation (100 mg/rat; SQ). The chronic pressor response to DOCA was attenuated in MnPOx rats by Day 11 of treatment and continued such that MAP increased 25 ± 3 mmHg in MnPOsham rats by Day 21 of DOCA compared to 14 ± 3 mmHg in MnPOx rats. These results support the hypothesis that the MnPO is an important brain site of action and necessary for the full development of DOCA-salt hypertension in the rat.


Assuntos
Acetato de Desoxicorticosterona , Hipertensão , Área Pré-Óptica , Ratos Sprague-Dawley , Animais , Masculino , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/metabolismo , Hipertensão/induzido quimicamente , Hipertensão/fisiopatologia , Hipertensão/etiologia , Ratos , Cloreto de Sódio na Dieta/efeitos adversos , Pressão Sanguínea/efeitos dos fármacos
8.
Peptides ; 177: 171226, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38649033

RESUMO

Close contact between lactating rodent mothers and their infants is essential for effective nursing. Whether the mother's effort to retrieve the infants to their nest requires the vasopressin-signaling via V1b receptor has not been fully defined. To address this question, V1b receptor knockout (V1bKO) and control mice were analyzed in pup retrieval test. Because an exploring mother in a new test cage randomly accessed to multiple infants in changing backgrounds over time, a computer vision-based deep learning analysis was applied to continuously calculate the distances between the mother and the infants as a parameter of their relationship. In an open-field, a virgin female V1bKO mice entered fewer times into the center area and moved shorter distances than wild-type (WT). While this behavioral pattern persisted in V1bKO mother, the pup retrieval test demonstrated that total distances between a V1bKO mother and infants came closer in a shorter time than with a WT mother. Moreover, in the medial preoptic area, parts of the V1b receptor transcripts were detected in galanin- and c-fos-positive neurons following maternal stimulation by infants. This research highlights the effectiveness of deep learning analysis in evaluating the mother-infant relationship and the critical role of V1b receptor in pup retrieval during the early lactation phase.


Assuntos
Comportamento Materno , Camundongos Knockout , Receptores de Vasopressinas , Animais , Feminino , Camundongos , Animais Recém-Nascidos , Aprendizado Profundo , Lactação/genética , Comportamento Materno/fisiologia , Área Pré-Óptica/metabolismo , Receptores de Vasopressinas/genética , Receptores de Vasopressinas/metabolismo
9.
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
10.
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
11.
Psychoneuroendocrinology ; 163: 106988, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38342055

RESUMO

Perinatal testosterone, or its metabolite estradiol, organize the brain toward a male phenotype. Male rodents with insufficient testosterone during this period fail to display sexual behavior and partner preference for receptive females in adulthood. However, cohabitation with non-reproductive conspecifics under the influence of a D2 agonist facilitates the expression of conditioned partner preference via Pavlovian learning in gonadally intact male rats. In the present experiment, three groups of neonatal PD1 males (N = 12/group) were either gonadectomized (GDX), sham-GDX, or left intact and evaluated for social preferences and sexual behaviors as adults. We then examined whether the effects of GDX could be reversed by conditioning the males via cohabitation with receptive females under the effects of the D2 agonist quinpirole (QNP) or saline, along with the size of some brain regions, such as the sexually dimorphic nucleus of the preoptic area (SDN-POA), suprachiasmatic nucleus (SCN), posterior dorsal medial amygdala (MeApd) and ventromedial hypothalamus (VMH). Results indicated that neonatal GDX resulted in the elimination of male-typical sexual behavior, an increase in same-sex social preference, and a reduction of the area of the SDN-POA. However, GDX-QNP males that underwent exposure to receptive females in adulthood increased their social preference for females and recovered the size in the SDN-POA. Although neonatal GDX impairs sexual behavior and disrupts partner preference and brain dimorphism in adult male rats, Pavlovian conditioning under enhanced D2 agonism ameliorates the effects on social preference and restores brain dimorphism in the SDN-POA without testosterone.


Assuntos
Área Pré-Óptica , Caracteres Sexuais , Gravidez , Ratos , Animais , Masculino , Feminino , Área Pré-Óptica/metabolismo , Encéfalo , Quimpirol/farmacologia , Castração , Testosterona/farmacologia , Testosterona/metabolismo
12.
BMC Neurosci ; 24(1): 58, 2023 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-37919674

RESUMO

The medial preoptic area (mPOA) regulates the probability and intensity of singing behavior in birds. Polzin and colleagues examined the molecular changes in the mPOA that were associated with gregarious song in European starlings (Sturnus vulgaris). High-throughput transcriptome analyses identified glutamate and dopamine pathways were highly enriched with gregarious song.


Assuntos
Estorninhos , Vocalização Animal , Animais , Vocalização Animal/fisiologia , Comportamento Sexual Animal/fisiologia , Comportamento Social , Estorninhos/metabolismo , Dopamina/metabolismo , Área Pré-Óptica/metabolismo
13.
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
14.
Cell Rep ; 42(9): 113029, 2023 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-37632750

RESUMO

The ventral tegmental area (VTA) has been proposed to play a role in pain, but the brain structures modulating VTA activity in response to nociceptive stimuli remain unclear. Here, we demonstrate that the lateral preoptic area (LPO) glutamate neurons relay nociceptive information to the VTA. These LPO glutamatergic neurons synapsing on VTA neurons respond to nociceptive stimulation and conditioned stimuli predicting nociceptive stimulation and also mediate aversion. In contrast, LPO GABA neurons synapsing in the VTA mediate reward. By ultrastructural quantitative synaptic analysis, ex vivo electrophysiology, and functional neuroanatomy we identify a complex circuitry between LPO glutamatergic and GABAergic neurons and VTA dopaminergic, GABAergic, and glutamatergic neurons. We conclude that LPO glutamatergic neurons play a causal role in the processing of nociceptive stimuli and in relaying information about nociceptive stimuli. The pathway from LPO glutamatergic neurons to the VTA represents an unpredicted interface between peripheral nociceptive information and the limbic system.


Assuntos
Ácido Glutâmico , Área Tegmentar Ventral , Ácido Glutâmico/metabolismo , Área Tegmentar Ventral/metabolismo , Área Pré-Óptica/metabolismo , Nociceptividade , Neurônios GABAérgicos/metabolismo , Neurônios Dopaminérgicos/metabolismo
15.
BMC Neurosci ; 24(1): 41, 2023 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-37537543

RESUMO

BACKGROUND: Song performed in flocks by European starlings (Sturnus vulgaris), referred to here as gregarious song, is a non-sexual, social behavior performed by adult birds. Gregarious song is thought to be an intrinsically reinforced behavior facilitated by a low-stress, positive affective state that increases social cohesion within a flock. The medial preoptic area (mPOA) is a region known to have a role in the production of gregarious song. However, the neurochemical systems that potentially act within this region to regulate song remain largely unexplored. In this study, we used RNA sequencing to characterize patterns of gene expression in the mPOA of male and female starlings singing gregarious song to identify possibly novel neurotransmitter, neuromodulator, and hormonal pathways that may be involved in the production of gregarious song. RESULTS: Differential gene expression analysis and rank rank hypergeometric analysis indicated that dopaminergic, cholinergic, and GABAergic systems were associated with the production of gregarious song, with multiple receptor genes (e.g., DRD2, DRD5, CHRM4, GABRD) upregulated in the mPOA of starlings who sang at high rates. Additionally, co-expression network analyses identified co-expressing gene clusters of glutamate signaling-related genes associated with song. One of these clusters contained five glutamate receptor genes and two glutamate scaffolding genes and was significantly enriched for genetic pathways involved in neurodevelopmental disorders associated with social deficits in humans. Two of these genes, GRIN1 and SHANK2, were positively correlated with performance of gregarious song. CONCLUSIONS: This work provides new insights into the role of the mPOA in non-sexual, gregarious song in starlings and highlights candidate genes that may play a role in gregarious social interactions across vertebrates. The provided data will also allow other researchers to compare across species to identify conserved systems that regulate social behavior.


Assuntos
Canto , Estorninhos , Animais , Humanos , Masculino , Feminino , Estorninhos/metabolismo , Vocalização Animal/fisiologia , Área Pré-Óptica/metabolismo , Expressão Gênica
16.
Nat Commun ; 14(1): 4924, 2023 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-37582782

RESUMO

Thermal homeostasis is vital for mammals and is controlled by brain neurocircuits. Yet, the neural pathways responsible for cold defense regulation are still unclear. Here, we found that a pathway from the lateral parabrachial nucleus (LPB) to the dorsomedial hypothalamus (DMH), which runs parallel to the canonical LPB to preoptic area (POA) pathway, is also crucial for cold defense. Together, these pathways make an equivalent and cumulative contribution, forming a parallel circuit. Specifically, activation of the LPB → DMH pathway induced strong cold-defense responses, including increases in thermogenesis of brown adipose tissue (BAT), muscle shivering, heart rate, and locomotion. Further, we identified somatostatin neurons in the LPB that target DMH to promote BAT thermogenesis. Therefore, we reveal a parallel circuit governing cold defense in mice, which enables resilience to hypothermia and provides a scalable and robust network in heat production, reshaping our understanding of neural circuit regulation of homeostatic behaviors.


Assuntos
Hipotermia , Termogênese , Camundongos , Animais , Termogênese/fisiologia , Área Pré-Óptica/metabolismo , Vias Neurais/fisiologia , Homeostase , Hipotermia/metabolismo , Tecido Adiposo Marrom/metabolismo , Temperatura Baixa , Mamíferos
17.
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
18.
Int J Mol Sci ; 24(14)2023 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-37511364

RESUMO

Male mammals display different paternal responses to pups, either attacking or killing the young offspring, or contrastingly, caring for them. The neural circuit mechanism underlying the between-individual variation in the pup-directed responsiveness of male mammals remains unclear. Monogamous mandarin voles were used to complete the present study. The male individuals were identified as paternal and infanticidal voles, according their behavioral responses to pups. It was found that the serotonin release in the medial preoptic area (MPOA), as well as the serotonergic neuron activity, significantly increased upon licking the pups, but showed no changes after attacking the pups, as revealed by the in vivo fiber photometry of the fluorescence signal from the 5-HT 1.0 sensor and the calcium imaging indicator, respectively. It was verified that the 5-HTergic neural projections to the MPOA originated mainly from the ventral part of the dorsal raphe (vDR). Furthermore, the chemogenetic inhibition of serotonergic projections from the vDR to the MPOA decreased the paternal behaviors and shortened the latency to attack the pups. In contrast, the activation of serotonergic neurons via optogenetics extended the licking duration and inhibited infanticide. Collectively, these results elucidate that the serotonergic projections from the vDR to the MPOA, a previously unrecognized pathway, regulate the paternal responses of virgin male mandarin voles to pups.


Assuntos
Núcleo Dorsal da Rafe , Área Pré-Óptica , Humanos , Animais , Masculino , Área Pré-Óptica/metabolismo , Pai , Comportamento Animal/fisiologia , Arvicolinae
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA