Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.710
Filtrar
1.
BMC Genomics ; 25(1): 771, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-39118023

RESUMEN

Prolonged or chronic social isolation has pronounced effects on animals, ranging from altered stress responses, increased anxiety and aggressive behaviour, and even increased mortality. The effects of shorter periods of isolation are much less well researched; however, short periods of isolation are used routinely for testing animal behaviour and physiology. Here, we studied how a 3 h period of isolation from a cagemate affected neural gene expression in three brain regions that contain important components of the social decision-making network, the hypothalamus, the nucleus taeniae of the amygdala, and the bed nucleus of the stria terminalis, using a gregarious bird as a model (zebra finches). We found evidence suggestive of altered neural activity, synaptic transmission, metabolism, and even potentially pain perception, all of which could create cofounding effects on experimental tests that involve isolating animals. We recommend that the effects of short-term social isolation need to be better understood and propose alternatives to isolating animals for testing.


Asunto(s)
Toma de Decisiones , Pinzones , Aislamiento Social , Animales , Aislamiento Social/psicología , Pinzones/fisiología , Masculino , Conducta Animal , Encéfalo/metabolismo , Encéfalo/fisiología , Núcleos Septales/metabolismo , Conducta Social , Amígdala del Cerebelo/metabolismo , Amígdala del Cerebelo/fisiología , Hipotálamo/metabolismo
2.
Cell Rep ; 43(7): 114489, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38990724

RESUMEN

It is well established that the basolateral amygdala (BLA) is an emotional processing hub that governs a diverse repertoire of behaviors. Selective engagement of a heterogeneous cell population in the BLA is thought to contribute to this flexibility in behavioral outcomes. However, whether this process is impacted by previous experiences that influence emotional processing remains unclear. Here we demonstrate that previous positive (enriched environment [EE]) or negative (chronic unpredictable stress [CUS]) experiences differentially influence the activity of populations of BLA principal neurons projecting to either the nucleus accumbens core or bed nucleus of the stria terminalis. Chemogenetic manipulation of these projection-specific neurons can mimic or occlude the effects of CUS and EE on behavioral outcomes to bidirectionally control avoidance behaviors and stress-induced helplessness. These data demonstrate that previous experiences influence the responsiveness of projection-specific BLA principal neurons, biasing information routing through the BLA, to drive divergent behavioral outcomes.


Asunto(s)
Complejo Nuclear Basolateral , Conducta Animal , Animales , Complejo Nuclear Basolateral/fisiología , Masculino , Neuronas/fisiología , Ratones , Estrés Psicológico , Núcleo Accumbens/fisiología , Ratones Endogámicos C57BL , Núcleos Septales/fisiología
3.
J Neurosci ; 44(32)2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39009438

RESUMEN

Neuroticism/negative emotionality (N/NE)-the tendency to experience anxiety, fear, and other negative emotions-is a fundamental dimension of temperament with profound consequences for health, wealth, and well-being. Elevated N/NE is associated with a panoply of adverse outcomes, from reduced socioeconomic attainment to psychiatric illness. Animal research suggests that N/NE reflects heightened reactivity to uncertain threat in the bed nucleus of the stria terminalis (BST) and central nucleus of the amygdala (Ce), but the relevance of these discoveries to humans has remained unclear. Here we used a novel combination of psychometric, psychophysiological, and neuroimaging approaches to test this hypothesis in an ethnoracially diverse, sex-balanced sample of 220 emerging adults selectively recruited to encompass a broad spectrum of N/NE. Cross-validated robust-regression analyses demonstrated that N/NE is preferentially associated with heightened BST activation during the uncertain anticipation of a genuinely distressing threat (aversive multimodal stimulation), whereas N/NE was unrelated to BST activation during certain-threat anticipation, Ce activation during either type of threat anticipation, or BST/Ce reactivity to threat-related faces. It is often assumed that different threat paradigms are interchangeable assays of individual differences in brain function, yet this has rarely been tested. Our results revealed negligible associations between BST/Ce reactivity to the anticipation of threat and the presentation of threat-related faces, indicating that the two tasks are nonfungible. These observations provide a framework for conceptualizing emotional traits and disorders; for guiding the design and interpretation of biobank and other neuroimaging studies of psychiatric risk, disease, and treatment; and for refining mechanistic research.


Asunto(s)
Amígdala del Cerebelo , Emociones , Imagen por Resonancia Magnética , Neuroticismo , Núcleos Septales , Núcleos Septales/fisiología , Núcleos Septales/diagnóstico por imagen , Humanos , Masculino , Femenino , Amígdala del Cerebelo/fisiología , Amígdala del Cerebelo/diagnóstico por imagen , Adulto Joven , Neuroticismo/fisiología , Adulto , Emociones/fisiología , Incertidumbre , Miedo/fisiología , Miedo/psicología , Adolescente
4.
Neurobiol Dis ; 199: 106596, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38986718

RESUMEN

Mesial temporal lobe epilepsy (MTLE) is characterized by recurring focal seizures that arise from limbic areas and are often refractory to pharmacological interventions. We have reported that optogenetic stimulation of PV-positive cells in the medial septum at 0.5 Hz exerts seizure-suppressive effects. Therefore, we compared here these results with those obtained by optogenetic stimulation of medial septum PV-positive neurons at 8 Hz in male PV-ChR2 mice (P60-P100) undergoing an initial, pilocarpine-induced status epilepticus (SE). Optogenetic stimulation (5 min ON, 10 min OFF) was performed from day 8 to day 12 after SE at a frequency of 8 Hz (n = 6 animals) or 0.5 Hz (n = 8 animals). Surprisingly, in both groups, no effects were observed on the occurrence of interictal spikes and interictal high frequency oscillations (HFOs). However, 0.5 Hz stimulation induced a significant decrease of seizure occurrence (p < 0.05). Such anti-ictogenic effect was not observed in the 8 Hz protocol that instead triggered seizures (p < 0.05); these seizures were significantly longer under optogenetic stimulation compared to when optogenetic stimulation was not implemented (p < 0.05). Analysis of ictal HFOs revealed that in the 0.5 Hz group, but not in the 8 Hz group, seizures occurring under optogenetic stimulation were associated with significantly lower rates of fast ripples compared to when optogenetic stimulation was not performed (p < 0.05). Our results indicate that activation of GABAergic PV-positive neurons in the medial septum exerts seizure-suppressing effects that are frequency-dependent and associated with low rates of fast ripples. Optogenetic activation of medial septum PV-positive neurons at 0.5 Hz is efficient in blocking seizures in the pilocarpine model of MTLE, an effect that did not occur with 8 Hz stimulation.


Asunto(s)
Epilepsia del Lóbulo Temporal , Optogenética , Convulsiones , Animales , Optogenética/métodos , Epilepsia del Lóbulo Temporal/fisiopatología , Epilepsia del Lóbulo Temporal/inducido químicamente , Epilepsia del Lóbulo Temporal/terapia , Masculino , Convulsiones/fisiopatología , Ratones , Pilocarpina/toxicidad , Ratones Transgénicos , Modelos Animales de Enfermedad , Tabique del Cerebro , Núcleos Septales/fisiopatología , Ratones Endogámicos C57BL
5.
Biol Psychiatry ; 95(8): 785-799, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38952926

RESUMEN

Background: Responding to social signals by expressing the correct behavior is not only challenged in autism, but also in diseases with high prevalence of autism, like Prader-Willi Syndrome (PWS). Clinical evidence suggests aberrant pro-social behavior in patients can be regulated by intranasal oxytocin (OXT) or vasopressin (AVP). However, what neuronal mechanisms underlie impaired behavioral responses in a socially-aversive context, and how can they be corrected, remains largely unknown. Methods: Using the Magel2 knocked-out (KO) mouse model of PWS (crossed with CRE-dependent transgenic lines), we devised optogenetic, physiological and pharmacological strategies in a social-fear-conditioning paradigm. Pathway specific roles of OXT and AVP signaling were investigated converging on the lateral septum (LS), a region which receives dense hypothalamic inputs. Results: OXT and AVP signaling promoted inhibitory synaptic transmission in the LS, which failure in Magel2KO mice disinhibited somatostatin (SST) neurons and disrupted social-fear extinction. The source of OXT and AVP deficits mapped specifically in the supraoptic nucleus→LS pathway of Magel2KO mice disrupting social-fear extinction, which could be corrected by optogenetic or pharmacological inhibition of SST-neurons in the LS. Interestingly, LS SST-neurons also gated the expression of aggressive behavior, possibly as part of functional units operating beyond local septal circuits. Conclusions: SST cells in the LS play a crucial role in integration and expression of disrupted neuropeptide signals in autism, thereby altering the balance in expression of safety versus fear. Our results uncover novel mechanisms underlying dysfunction in a socially-aversive context, and provides a new framework for future treatments in autism-spectrum disorders.


Asunto(s)
Modelos Animales de Enfermedad , Extinción Psicológica , Miedo , Ratones Noqueados , Neuronas , Oxitocina , Síndrome de Prader-Willi , Somatostatina , Vasopresinas , Animales , Oxitocina/farmacología , Somatostatina/farmacología , Somatostatina/metabolismo , Miedo/efectos de los fármacos , Miedo/fisiología , Extinción Psicológica/efectos de los fármacos , Extinción Psicológica/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratones , Síndrome de Prader-Willi/fisiopatología , Síndrome de Prader-Willi/tratamiento farmacológico , Vasopresinas/metabolismo , Agresión/efectos de los fármacos , Agresión/fisiología , Masculino , Conducta Social , Núcleos Septales/efectos de los fármacos , Núcleos Septales/metabolismo , Optogenética , Ratones Endogámicos C57BL , Péptidos y Proteínas de Señalización Intracelular , Proteínas Intrínsecamente Desordenadas
6.
Nat Commun ; 15(1): 5805, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38987240

RESUMEN

Fear memory is essential for survival and adaptation, yet excessive fear memories can lead to emotional disabilities and mental disorders. Despite previous researches have indicated that histamine H1 receptor (H1R) exerts critical and intricate effects on fear memory, the role of H1R is still not clarified. Here, we show that deletion of H1R gene in medial septum (MS) but not other cholinergic neurons selectively enhances contextual fear memory without affecting cued memory by differentially activating the dentate gyrus (DG) neurons in mice. H1R in cholinergic neurons mediates the contextual fear retrieval rather than consolidation by decreasing acetylcholine release pattern in DG. Furthermore, selective knockdown of H1R in the MS is sufficient to enhance contextual fear memory by manipulating the retrieval-induced neurons in DG. Our results suggest that H1R in MS cholinergic neurons is critical for contextual fear retrieval, and could be a potential therapeutic target for individuals with fear-related disorders.


Asunto(s)
Neuronas Colinérgicas , Giro Dentado , Miedo , Receptores Histamínicos H1 , Animales , Miedo/fisiología , Neuronas Colinérgicas/metabolismo , Neuronas Colinérgicas/fisiología , Receptores Histamínicos H1/metabolismo , Receptores Histamínicos H1/genética , Giro Dentado/metabolismo , Ratones , Masculino , Ratones Endogámicos C57BL , Memoria/fisiología , Ratones Noqueados , Acetilcolina/metabolismo , Núcleos Septales/metabolismo , Núcleos Septales/fisiología , Núcleos Septales/citología
7.
Cell Rep ; 43(6): 114343, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38865247

RESUMEN

Activation of prepronociceptin (PNOC)-expressing neurons in the arcuate nucleus (ARC) promotes high-fat-diet (HFD)-induced hyperphagia. In turn, PNOCARC neurons can inhibit the anorexic response of proopiomelanocortin (POMC) neurons. Here, we validate the necessity of PNOCARC activity for HFD-induced inhibition of POMC neurons in mice and find that PNOCARC-neuron-dependent inhibition of POMC neurons is mediated by gamma-aminobutyric acid (GABA) release. When monitoring individual PNOCARC neuron activity via Ca2+ imaging, we find a subpopulation of PNOCARC neurons that is inhibited upon gastrointestinal calorie sensing and disinhibited upon HFD feeding. Combining retrograde rabies tracing and circuit mapping, we find that PNOC neurons from the bed nucleus of the stria terminalis (PNOCBNST) provide inhibitory input to PNOCARC neurons, and this inhibitory input is blunted upon HFD feeding. This work sheds light on how an increase in caloric content of the diet can rewire a neuronal circuit, paving the way to overconsumption and obesity development.


Asunto(s)
Dieta Alta en Grasa , Hiperfagia , Núcleos Septales , Animales , Hiperfagia/metabolismo , Ratones , Núcleos Septales/metabolismo , Neuronas/metabolismo , Masculino , Ácido gamma-Aminobutírico/metabolismo , Proopiomelanocortina/metabolismo , Neuronas GABAérgicas/metabolismo , Núcleo Arqueado del Hipotálamo/metabolismo , Ratones Endogámicos C57BL , Precursores de Proteínas , Receptores Opioides
8.
Behav Brain Res ; 471: 115116, 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-38897419

RESUMEN

The neural mechanisms underlying paternal care in biparental mammals are not well understood. The California mouse (Peromyscus californicus) is a biparental rodent in which virtually all fathers are attracted to pups, while virgin males vary widely in their behavior toward unrelated infants, ranging from attacking to avoiding to huddling and grooming pups. We previously showed that pharmacologically inhibiting the synthesis of the neurotransmitter norepinephrine (NE) with the dopamine ß-hydroxylase inhibitor nepicastat reduced the propensity of virgin male and female California mice to interact with pups. The current study tested the hypothesis that nepicastat would reduce pup-induced c-Fos immunoreactivity, a cellular marker of neural activity, in the medial preoptic area (MPOA), medial amygdala (MeA), basolateral amygdala (BLA), and bed nucleus of the stria terminalis (BNST), brain regions implicated in the control of parental behavior and/or anxiety. Virgin males were injected with nepicastat (75 mg/kg, i.p.) or vehicle 2 hours prior to exposure to either an unrelated pup or novel object for 60 minutes (n = 4-6 mice per group). Immediately following the 60-minute stimulus exposure, mice were euthanized and their brains were collected for c-Fos immunohistochemistry. Nepicastat reduced c-Fos expression in the MeA and MPOA of pup-exposed virgin males compared to vehicle-injected controls. In contrast, nepicastat did not alter c-Fos expression in any of the above brain regions following exposure to a novel object. Overall, these results suggest that the noradrenergic system might influence MeA and MPOA function to promote behavioral interactions with pups in virgin males.


Asunto(s)
Dopamina beta-Hidroxilasa , Conducta Paterna , Peromyscus , Área Preóptica , Núcleos Septales , Animales , Masculino , Dopamina beta-Hidroxilasa/metabolismo , Dopamina beta-Hidroxilasa/antagonistas & inhibidores , Conducta Paterna/fisiología , Conducta Paterna/efectos de los fármacos , Núcleos Septales/efectos de los fármacos , Núcleos Septales/metabolismo , Área Preóptica/metabolismo , Área Preóptica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Femenino , Inhibidores Enzimáticos/farmacología , Complejo Nuclear Basolateral/efectos de los fármacos , Complejo Nuclear Basolateral/metabolismo , Complejo Nuclear Corticomedial/efectos de los fármacos , Complejo Nuclear Corticomedial/metabolismo , Norepinefrina/metabolismo , Imidazoles , Tionas
9.
Neuropharmacology ; 257: 110033, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-38866066

RESUMEN

The anteroventral bed nucleus of stria terminalis (avBNST) is a limbic forebrain region involved in the regulation of anxiety, and expresses GABAB receptors, which are located at both pre- and post-synaptic sites. However, it is unclear how blockade of these receptors affects anxiety-like behaviors, particularly in Parkinson's disease (PD)-related anxiety. In the present study, unilateral 6-hydroxydopamine (6-OHDA) lesions of the substantia nigra pars compacta in rats induced anxiety-like behaviors, and increased GABA release and decreased glutamate release in the avBNST, as well as decreased level of dopamine (DA) in the basolateral amygdala (BLA). Intra-avBNST injection of pre-synaptic GABAB receptor antagonist CGP36216 produced anxiolytic-like effects, while the injection of post-synaptic GABAB receptor antagonist CGP35348 induced anxiety-like responses in both sham and 6-OHDA rats. Intra-avBNST injection of CGP36216 inhibited the GABAergic neurons and increased GABA/glutamate ratio in the avBNST and increased levels of DA and serotonin (5-HT) in the BLA; conversely, CGP35348 produced opposite effects on the firing activity of avBNST GABAergic neurons and levels of the neurotransmitters in the avBNST and BLA. Moreover, the doses of the antagonists producing significant behavioral effects in 6-OHDA rats were lower than those in sham rats, and the duration of action of the antagonists on the firing rate of the neurons and release of the neurotransmitters was prolonged in 6-OHDA rats. Altogether, these findings suggest that pre- and post-synaptic GABAB receptors in the avBNST are implicated in PD-related anxiety-like behaviors, and degeneration of the nigrostriatal pathway enhances functions and/or upregulates expression of these receptors.


Asunto(s)
Ansiolíticos , Ansiedad , Antagonistas de Receptores de GABA-B , Oxidopamina , Trastornos Parkinsonianos , Receptores de GABA-B , Núcleos Septales , Animales , Núcleos Septales/efectos de los fármacos , Núcleos Septales/metabolismo , Masculino , Ansiedad/metabolismo , Antagonistas de Receptores de GABA-B/farmacología , Ansiolíticos/farmacología , Ratas , Receptores de GABA-B/metabolismo , Oxidopamina/toxicidad , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/psicología , Dopamina/metabolismo , Ácido Glutámico/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Ratas Sprague-Dawley , Serotonina/metabolismo , Complejo Nuclear Basolateral/metabolismo , Complejo Nuclear Basolateral/efectos de los fármacos , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/metabolismo , Compuestos Organofosforados
10.
J Physiol ; 602(14): 3545-3574, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38874572

RESUMEN

Melanin-concentrating hormone (MCH) neurons can co-express several neuropeptides or neurotransmitters and send widespread projections throughout the brain. Notably, there is a dense cluster of nerve terminals from MCH neurons in the lateral septum (LS) that innervate LS cells by glutamate release. The LS is also a key region integrating stress- and anxiety-like behaviours, which are also emerging roles of MCH neurons. However, it is not known if or where the MCH peptide acts within the LS. We analysed the projections from MCH neurons in male and female mice anteroposteriorly throughout the LS and found spatial overlap between the distribution pattern of MCH-immunoreactive (MCH-ir) fibres with MCH receptor Mchr1 mRNA hybridization or MCHR1-ir cells. This overlap was most prominent along the ventral and lateral border of the rostral part of the LS (LSr). Most MCHR1-labelled LS neurons lay adjacent to passing MCH-ir fibres, but some MCH-ir varicosities directly contacted the soma or cilium of MCHR1-labelled LS neurons. We thus performed whole-cell patch-clamp recordings from MCHR1-rich LSr regions to determine if and how LS cells respond to MCH. Bath application of MCH to acute brain slices activated a bicuculline-sensitive chloride current that directly hyperpolarized LS cells. This MCH-mediated hyperpolarization was blocked by calphostin C, which suggested that the inhibitory actions of MCH were mediated by protein kinase C-dependent activation of GABAA receptors. Taken together, these findings define potential hotspots within the LS that may elucidate the contributions of MCH to stress- or anxiety-related feeding behaviours. KEY POINTS: Melanin-concentrating hormone (MCH) neurons have dense nerve terminals within the lateral septum (LS), a key region underlying stress- and anxiety-like behaviours that are emerging roles of the MCH system, but the function of MCH in the LS is not known. We found spatial overlap between MCH-immunoreactive fibres, Mchr1 mRNA, and MCHR1 protein expression along the lateral border of the LS. Within MCHR1-rich regions, MCH directly inhibited LS cells by increasing chloride conductance via GABAA receptor activation in a protein kinase C-dependent manner. Electrophysiological MCH effects in brain slices have been elusive, and few studies have described the mechanisms of MCH action. Our findings demonstrated, to our knowledge, the first description of MCHR1 Gq-coupling in brain slices, which was previously predicted in cell or primary culture models only. Together, these findings defined hotspots and mechanistic underpinnings for MCH effects such as in feeding and anxiety-related behaviours.


Asunto(s)
Hormonas Hipotalámicas , Melaninas , Neuronas , Hormonas Hipofisarias , Receptores de Somatostatina , Núcleos Septales , Animales , Hormonas Hipotalámicas/metabolismo , Melaninas/metabolismo , Hormonas Hipofisarias/metabolismo , Masculino , Femenino , Ratones , Núcleos Septales/metabolismo , Núcleos Septales/fisiología , Receptores de Somatostatina/metabolismo , Neuronas/metabolismo , Neuronas/fisiología , Ratones Endogámicos C57BL
11.
Neurosci Lett ; 834: 137848, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38823510

RESUMEN

Ketamine is a dissociative anesthetic that has been proposed to be a useful alternative in cases of a poor response to other treatments in patients with depression. Remarkably, beneficial clinical actions of ketamine are detected once its psychotropic actions disappear. Therefore, clinical actions may occur independently of dose. Most current studies focus on actions of ketamine on neurotrophic factors, but few studies have investigated actions of ketamine on neural structures for which actions of antidepressants have been previously explored. Lateral septal nucleus (LSN) stimulation reduces neural activity in the prelimbic cortex (PL) and infralimbic cortex (IL) subregions of the medial prefrontal cortex (mPFC). Fluoxetine increases inhibitory responsivity of the LSN-IL connection. In the present study, actions of an anesthetic dose of ketamine were compared with a high dose of fluoxetine on behavior and neural responsivity 24 h after drug administration. Fluoxetine reduced immobility in the forced swim test without changing locomotor activity in the open field test. Ketamine strongly decreased locomotor activity and did not produce changes in immobility. In another set of Wistar rats that received similar drug treatment regimens, the results indicated that LSN stimulation in saline-treated animals produced a long-lasting inhibitory afterdischarge in these mPFC subregions. Actions of ketamine on the LSN-mPFC connection reproduced actions of fluoxetine, consisting of accentuated inhibition of the LSN action on the mPFC. These findings suggest that independent of different actions on neurotransmission, the common final pathway of antidepressants lies in their actions on forebrain structures that are related to emotional regulation.


Asunto(s)
Fluoxetina , Ketamina , Corteza Prefrontal , Ratas Wistar , Núcleos Septales , Animales , Ketamina/farmacología , Fluoxetina/farmacología , Masculino , Corteza Prefrontal/efectos de los fármacos , Ratas , Núcleos Septales/efectos de los fármacos , Estimulación Eléctrica
12.
Nat Commun ; 15(1): 5439, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38937485

RESUMEN

Efficient control of feeding behavior requires the coordinated adjustment of complex motivational and affective neurocircuits. Neuropeptides from energy-sensing hypothalamic neurons are potent feeding modulators, but how these endogenous signals shape relevant circuits remains unclear. Here, we examine how the orexigenic neuropeptide Y (NPY) adapts GABAergic inputs to the bed nucleus of the stria terminalis (BNST). We find that fasting increases synaptic connectivity between agouti-related peptide (AgRP)-expressing 'hunger' and BNST neurons, a circuit that promotes feeding. In contrast, GABAergic input from the central amygdala (CeA), an extended amygdala circuit that decreases feeding, is reduced. Activating NPY-expressing AgRP neurons evokes these synaptic adaptations, which are absent in NPY-deficient mice. Moreover, fasting diminishes the ability of CeA projections in the BNST to suppress food intake, and NPY-deficient mice fail to decrease anxiety in order to promote feeding. Thus, AgRP neurons drive input-specific synaptic plasticity, enabling a selective shift in hunger and anxiety signaling during starvation through NPY.


Asunto(s)
Proteína Relacionada con Agouti , Conducta Alimentaria , Plasticidad Neuronal , Neuropéptido Y , Núcleos Septales , Inanición , Animales , Neuropéptido Y/metabolismo , Neuropéptido Y/genética , Plasticidad Neuronal/fisiología , Proteína Relacionada con Agouti/metabolismo , Proteína Relacionada con Agouti/genética , Conducta Alimentaria/fisiología , Núcleos Septales/metabolismo , Núcleos Septales/fisiología , Ratones , Inanición/metabolismo , Masculino , Amígdala del Cerebelo/metabolismo , Amígdala del Cerebelo/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Neuronas/fisiología , Neuronas GABAérgicas/metabolismo , Ingestión de Alimentos/fisiología , Ayuno/fisiología , Ansiedad/metabolismo , Ansiedad/fisiopatología , Hambre/fisiología
13.
Theranostics ; 14(7): 2881-2896, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38773977

RESUMEN

Methamphetamine (METH) withdrawal anxiety symptom and relapse have been significant challenges for clinical practice, however, the underlying neuronal basis remains unclear. Our recent research has identified a specific subpopulation of choline acetyltransferase (ChAT+) neurons localized in the external lateral portion of parabrachial nucleus (eLPBChAT), which modulates METH primed-reinstatement of conditioned place preference (CPP). Here, the anatomical structures and functional roles of eLPBChAT projections in METH withdrawal anxiety and primed reinstatement were further explored. Methods: In the present study, a multifaceted approach was employed to dissect the LPBChAT+ projections in male mice, including anterograde and retrograde tracing, acetylcholine (Ach) indicator combined with fiber photometry recording, photogenetic and chemogenetic regulation, as well as electrophysiological recording. METH withdrawal anxiety-like behaviors and METH-primed reinstatement of conditioned place preference (CPP) were assessed in male mice. Results: We identified that eLPBChAT send projections to PKCδ-positive (PKCδ+) neurons in lateral portion of central nucleus of amygdala (lCeAPKCδ) and oval portion of bed nucleus of the stria terminalis (ovBNSTPKCδ), forming eLPBChAT-lCeAPKCδ and eLPBChAT-ovBNSTPKCδ pathways. At least in part, the eLPBChAT neurons positively innervate lCeAPKCδ neurons and ovBNSTPKCδ neurons through regulating synaptic elements of presynaptic Ach release and postsynaptic nicotinic acetylcholine receptors (nAChRs). METH withdrawal anxiety and METH-primed reinstatement of CPP respectively recruit eLPBChAT-lCeAPKCδ pathway and eLPBChAT-ovBNSTPKCδ pathway in male mice. Conclusion: Our findings put new insights into the complex neural networks, especially focusing on the eLPBChAT projections. The eLPBChAT is a critical node in the neural networks governing METH withdrawal anxiety and primed-reinstatement of CPP through its projections to the lCeAPKCδ and ovBNSTPKCδ, respectively.


Asunto(s)
Ansiedad , Metanfetamina , Ratones Endogámicos C57BL , Síndrome de Abstinencia a Sustancias , Animales , Metanfetamina/efectos adversos , Masculino , Ratones , Síndrome de Abstinencia a Sustancias/metabolismo , Síndrome de Abstinencia a Sustancias/fisiopatología , Ansiedad/metabolismo , Neuronas/metabolismo , Colina O-Acetiltransferasa/metabolismo , Núcleos Septales/metabolismo , Conducta Animal/efectos de los fármacos
14.
Psychoneuroendocrinology ; 166: 107083, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38788461

RESUMEN

In mammals, some physiological conditions are associated with the high brain oxytocin (OXT) system activity. These include lactation in females and mating in males and females, both of which have been linked to reduced stress responsiveness and anxiolysis. Also, in a murine model of social fear conditioning (SFC), enhanced brain OXT signaling in lactating mice, specifically in the lateral septum (LS), was reported to underlie reduced social fear expression. Here, we studied the effects of mating in male mice on anxiety-related behaviour, social (and cued) fear expression and its extinction, and the activity of OXT neurons reflected by cFos expression and OXT release in the LS and amygdala. We further focused on the involvement of brain OXT in the mating-induced facilitation of social fear extinction. We could confirm the anxiolytic effect of mating in male mice irrespective of the occurrence of ejaculation. Further, we found that only successful mating resulting in ejaculation (Ej+) facilitated social fear extinction, whereas mating without ejaculation (Ej-) did not. In contrast, mating did not affect cues fear expression. Using the cellular activity markers cFos and pErk, we further identified the ventral LS (vLS) as a potential region participating in the effect of ejaculation on social fear extinction. In support, microdialysis experiments revealed a rise in OXT release within the LS, but not the amygdala, during mating. Finally, infusion of an OXT receptor antagonist into the LS before mating or into the lateral ventricle (icv) after mating demonstrated a significant role of brain OXT receptor-mediated signaling in the mating-induced facilitation of social fear extinction.


Asunto(s)
Amígdala del Cerebelo , Extinción Psicológica , Miedo , Oxitocina , Conducta Sexual Animal , Animales , Miedo/fisiología , Oxitocina/metabolismo , Masculino , Extinción Psicológica/fisiología , Ratones , Femenino , Conducta Sexual Animal/fisiología , Amígdala del Cerebelo/metabolismo , Conducta Social , Ansiedad/metabolismo , Receptores de Oxitocina/metabolismo , Núcleos Septales/metabolismo , Núcleos Septales/efectos de los fármacos , Eyaculación/fisiología , Copulación/fisiología , Tabique del Cerebro/metabolismo , Tabique del Cerebro/fisiología , Ratones Endogámicos C57BL , Conducta Animal/fisiología , Conducta Animal/efectos de los fármacos
16.
Proc Natl Acad Sci U S A ; 121(20): e2319641121, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38709918

RESUMEN

One of the largest sex differences in brain neurochemistry is the expression of the neuropeptide arginine vasopressin (AVP) within the vertebrate brain, with males having more AVP cells in the bed nucleus of the stria terminalis (BNST) than females. Despite the long-standing implication of AVP in social and anxiety-like behaviors, the circuitry underlying AVP's control of these behaviors is still not well defined. Using optogenetic approaches, we show that inhibiting AVP BNST cells reduces social investigation in males, but not in females, whereas stimulating these cells increases social investigation in both sexes, but more so in males. These cells may facilitate male social investigation through their projections to the lateral septum (LS), an area with the highest density of sexually differentiated AVP innervation in the brain, as optogenetic stimulation of BNST AVP → LS increased social investigation and anxiety-like behavior in males but not in females; the same stimulation also caused a biphasic response of LS cells ex vivo. Blocking the vasopressin 1a receptor (V1aR) in the LS eliminated all these responses. Together, these findings establish a sexually differentiated role for BNST AVP cells in the control of social investigation and anxiety-like behavior, likely mediated by their projections to the LS.


Asunto(s)
Ansiedad , Arginina Vasopresina , Conducta Social , Animales , Femenino , Masculino , Ratones , Ansiedad/metabolismo , Arginina Vasopresina/metabolismo , Conducta Animal/fisiología , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuronas/fisiología , Optogenética , Receptores de Vasopresinas/metabolismo , Receptores de Vasopresinas/genética , Núcleos Septales/metabolismo , Núcleos Septales/fisiología
17.
Mol Brain ; 17(1): 22, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702738

RESUMEN

We previously reported that enhanced corticotropin-releasing factor (CRF) signaling in the bed nucleus of the stria terminalis (BNST) caused the aversive responses during acute pain and suppressed the brain reward system during chronic pain. However, it remains to be examined whether chronic pain alters the excitability of CRF neurons in the BNST. In this study we investigated the chronic pain-induced changes in excitability of CRF-expressing neurons in the oval part of the BNST (ovBNSTCRF neurons) by whole-cell patch-clamp electrophysiology. CRF-Cre; Ai14 mice were used to visualize CRF neurons by tdTomato. Electrophysiological recordings from brain slices prepared from a mouse model of neuropathic pain revealed that rheobase and firing threshold were significantly decreased in the chronic pain group compared with the sham-operated control group. Firing rate of the chronic pain group was higher than that of the control group. These data indicate that chronic pain elevated neuronal excitability of ovBNSTCRF neurons.


Asunto(s)
Dolor Crónico , Hormona Liberadora de Corticotropina , Neuronas , Núcleos Septales , Animales , Núcleos Septales/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Neuronas/metabolismo , Dolor Crónico/fisiopatología , Dolor Crónico/metabolismo , Masculino , Potenciales de Acción/fisiología , Ratones Endogámicos C57BL , Ratones
18.
Biochem Biophys Res Commun ; 721: 150145, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-38795633

RESUMEN

Itch, a common somatic sensation, serves as a crucial protective system. Recent studies have unraveled the neural mechanisms of itch at peripheral, spinal cord as well as cerebral levels. However, a comprehensive understanding of the central mechanism governing itch transmission and regulation remains elusive. Here, we report the role of the medial septum (MS), an integral component of the basal forebrain, in modulating the acute itch processing. The increases in c-Fos+ neurons and calcium signals within the MS during acute itch processing were observed. Pharmacogenetic activation manipulation of global MS neurons suppressed the scratching behaviors induced by chloroquine or compound 48/80. Microinjection of GABA into the MS or pharmacogenetic inhibition of non-GABAergic neurons markedly suppressed chloroquine-induced scratching behaviors. Pharmacogenetic activation of the MS-ACC GABAergic pathway attenuated chloroquine-induced acute itch. Hence, our findings reveal that MS has a regulatory role in the chloroquine-induced acute itch through local increased GABA to inhibit non-GABAergic neurons and the activation of MS-ACC GABAergic pathway.


Asunto(s)
Cloroquina , Giro del Cíngulo , Prurito , Ácido gamma-Aminobutírico , Cloroquina/farmacología , Animales , Prurito/inducido químicamente , Prurito/metabolismo , Prurito/tratamiento farmacológico , Masculino , Ácido gamma-Aminobutírico/metabolismo , Giro del Cíngulo/metabolismo , Giro del Cíngulo/efectos de los fármacos , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones , Núcleos Septales/metabolismo , Núcleos Septales/efectos de los fármacos
19.
Sci Adv ; 10(19): eadk7636, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38728397

RESUMEN

Corticotropin releasing factor (CRF) network in the oval nucleus of bed nuclei of the stria terminalis (ovBNST) is generally indicated in stress, but its role in female-biased susceptibility to anxiety is unknown. Here, we established a female-biased stress paradigm. We found that the CRF release in ovBNST during stress showed female-biased pattern, and ovBNST CRF neurons were more prone to be hyperexcited in female mice during stress in both in vitro and in vivo studies. Moreover, optogenetic modulation to exchange the activation pattern of ovBNST CRF neurons during stress between female and male mice could reverse their susceptibility to anxiety. Last, CRF receptor type 1 (CRFR1) mediated the CRF-induced excitation of ovBNST CRF neurons and showed female-biased expression. Specific knockdown of the CRFR1 level in ovBNST CRF neurons in female or overexpression that in male could reverse their susceptibility to anxiety. Therefore, we identify that CRFR1-mediated hyperexcitation of ovBNST CRF neurons in female mice encode the female-biased susceptibility to anxiety.


Asunto(s)
Ansiedad , Hormona Liberadora de Corticotropina , Neuronas , Receptores de Hormona Liberadora de Corticotropina , Animales , Femenino , Masculino , Ratones , Ansiedad/metabolismo , Reacción de Prevención/fisiología , Conducta Animal , Hormona Liberadora de Corticotropina/metabolismo , Neuronas/metabolismo , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Receptores de Hormona Liberadora de Corticotropina/genética , Núcleos Septales/metabolismo , Estrés Psicológico/metabolismo
20.
Sci Rep ; 14(1): 8919, 2024 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-38637645

RESUMEN

The natural alignment of animals into social dominance hierarchies produces adaptive, and potentially maladaptive, changes in the brain that influence health and behavior. Aggressive and submissive behaviors assumed by animals through dominance interactions engage stress-dependent neural and hormonal systems that have been shown to correspond with social rank. Here, we examined the association between social dominance hierarchy status established within cages of group-housed mice and the expression of the stress peptide PACAP in the bed nucleus of the stria terminalis (BNST) and central nucleus of the amygdala (CeA). We also examined the relationship between social dominance rank and blood corticosterone (CORT) levels, body weight, motor coordination (rotorod) and acoustic startle. Male C57BL/6 mice were ranked as either Dominant, Submissive, or Intermediate based on counts of aggressive/submissive encounters assessed at 12 weeks-old following a change in homecage conditions. PACAP expression was significantly higher in the BNST, but not the CeA, of Submissive mice compared to the other groups. CORT levels were lowest in Submissive mice and appeared to reflect a blunted response following events where dominance status is recapitulated. Together, these data reveal changes in specific neural/neuroendocrine systems that are predominant in animals of lowest social dominance rank, and implicate PACAP in brain adaptations that occur through the development of social dominance hierarchies.


Asunto(s)
Corticosterona , Núcleos Septales , Animales , Masculino , Ratones , Amígdala del Cerebelo/metabolismo , Ratones Endogámicos C57BL , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Núcleos Septales/metabolismo , Predominio Social , Estrés Psicológico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA