Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 733
Filtrar
1.
Nature ; 626(7998): 347-356, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38267576

RESUMEN

To survive in a complex social group, one needs to know who to approach and, more importantly, who to avoid. In mice, a single defeat causes the losing mouse to stay away from the winner for weeks1. Here through a series of functional manipulation and recording experiments, we identify oxytocin neurons in the retrochiasmatic supraoptic nucleus (SOROXT) and oxytocin-receptor-expressing cells in the anterior subdivision of the ventromedial hypothalamus, ventrolateral part (aVMHvlOXTR) as a key circuit motif for defeat-induced social avoidance. Before defeat, aVMHvlOXTR cells minimally respond to aggressor cues. During defeat, aVMHvlOXTR cells are highly activated and, with the help of an exclusive oxytocin supply from the SOR, potentiate their responses to aggressor cues. After defeat, strong aggressor-induced aVMHvlOXTR cell activation drives the animal to avoid the aggressor and minimizes future defeat. Our study uncovers a neural process that supports rapid social learning caused by defeat and highlights the importance of the brain oxytocin system in social plasticity.


Asunto(s)
Agresión , Reacción de Prevención , Hipotálamo , Vías Nerviosas , Neuronas , Oxitocina , Aprendizaje Social , Animales , Ratones , Agresión/fisiología , Reacción de Prevención/fisiología , Señales (Psicología) , Miedo/fisiología , Hipotálamo/citología , Hipotálamo/metabolismo , Vías Nerviosas/fisiología , Neuronas/metabolismo , Oxitocina/metabolismo , Receptores de Oxitocina/metabolismo , Conducta Social , Aprendizaje Social/fisiología , Núcleo Supraóptico/citología , Núcleo Supraóptico/metabolismo , Núcleo Hipotalámico Ventromedial/citología , Núcleo Hipotalámico Ventromedial/metabolismo , Plasticidad Neuronal
2.
Neurochem Res ; 46(10): 2586-2600, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33216313

RESUMEN

Studies on the interactions between astrocytes and neurons in the hypothalamo-neurohypophysial system have significantly facilitated our understanding of the regulation of neural activities. This has been exemplified in the interactions between astrocytes and magnocellular neuroendocrine cells (MNCs) in the supraoptic nucleus (SON), specifically during osmotic stimulation and lactation. In response to changes in neurochemical environment in the SON, astrocytic morphology and functions change significantly, which further modulates MNC activity and the secretion of vasopressin and oxytocin. In osmotic regulation, short-term dehydration or water overload causes transient retraction or expansion of astrocytic processes, which increases or decreases the activity of SON neurons, respectively. Prolonged osmotic stimulation causes adaptive change in astrocytic plasticity in the SON, which allows osmosensory neurons to reserve osmosensitivity at new levels. During lactation, changes in neurochemical environment cause retraction of astrocytic processes around oxytocin neurons, which increases MNC's ability to secrete oxytocin. During suckling by a baby/pup, astrocytic processes in the mother/dams exhibit alternative retraction and expansion around oxytocin neurons, which mirrors intermittently synchronized activation of oxytocin neurons and the post-excitation inhibition, respectively. The morphological and functional plasticities of astrocytes depend on a series of cellular events involving glial fibrillary acidic protein, aquaporin 4, volume regulated anion channels, transporters and other astrocytic functional molecules. This review further explores mechanisms underlying astroglial regulation of the neuroendocrine neuronal activities in acute processes based on the knowledge from studies on the SON.


Asunto(s)
Astrocitos/metabolismo , Células Neuroendocrinas/metabolismo , Núcleo Supraóptico/metabolismo , Animales , Acuaporina 4/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Lactancia/fisiología , Plasticidad Neuronal/fisiología , Osmorregulación/fisiología , Núcleo Supraóptico/citología
3.
Horm Behav ; 122: 104734, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32169583

RESUMEN

Oxytocin regulates social behaviours, pair bonding and hippocampal neurogenesis but most studies have used adult males. Our study investigated the effects of oxytocin on social investigation and adult hippocampal neurogenesis in male and female rats. Oxytocin has poor penetration of the blood-brain barrier, therefore we tested a nanoparticle drug, TRIOZAN™ (Ovensa Inc.), which permits greater blood-brain-barrier penetration. Adult male and female rats were injected daily (i.p.) for 10 days with either: oxytocin in PBS (0.5 or 1.0 mg/kg), oxytocin in TRIOZAN™ (0.5 or 1.0 mg/kg), or vehicle (PBS) and tested for social investigation. Oxytocin decreased body mass and increased social investigation and number of oxytocin-immunoreactive cells in the supraoptic nucleus (SON) of the hypothalamus in male rats only. In both sexes, oxytocin decreased the number of immature neurons (doublecortin+ cells) in the ventral hippocampus and reduced plasma 17ß-estradiol levels in a dose- and delivery-dependent way. Oxytocin in TRIOZAN™ reduced "sedation" observed post-injection and increased certain central effects (oxytocin levels in the hypothalamus and neurogenesis in the ventral hippocampus) relative to oxytocin in PBS, indicating that the nanoparticle may be used as an alternative brain delivery system. We showed that oxytocin has sex-specific effects on social investigation, body mass, "sedation", and the oxytocin system. In contrast, similar effects were observed in both sexes in neurogenesis and plasma 17ß-estradiol. Our work suggests that sex differences in oxytocin regulation of brain endpoints is region-specific (hypothalamus versus hippocampus) and that oxytocin does not promote social investigation in females.


Asunto(s)
Hipocampo/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Oxitocina/farmacología , Conducta Social , Animales , Conducta Animal/efectos de los fármacos , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Femenino , Hipocampo/citología , Hipocampo/fisiología , Hipotálamo/citología , Hipotálamo/metabolismo , Inmunohistoquímica , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Neuropéptidos/metabolismo , Oxitocina/metabolismo , Ratas , Ratas Sprague-Dawley , Caracteres Sexuales , Núcleo Supraóptico/citología , Núcleo Supraóptico/efectos de los fármacos , Núcleo Supraóptico/metabolismo
4.
Neuron ; 102(5): 1053-1065.e4, 2019 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-31006556

RESUMEN

How general anesthesia (GA) induces loss of consciousness remains unclear, and whether diverse anesthetic drugs and sleep share a common neural pathway is unknown. Previous studies have revealed that many GA drugs inhibit neural activity through targeting GABA receptors. Here, using Fos staining, ex vivo brain slice recording, and in vivo multi-channel electrophysiology, we discovered a core ensemble of hypothalamic neurons in and near the supraoptic nucleus, consisting primarily of neuroendocrine cells, which are persistently and commonly activated by multiple classes of GA drugs. Remarkably, chemogenetic or brief optogenetic activations of these anesthesia-activated neurons (AANs) strongly promote slow-wave sleep and potentiates GA, whereas conditional ablation or inhibition of AANs led to diminished slow-wave oscillation, significant loss of sleep, and shortened durations of GA. These findings identify a common neural substrate underlying diverse GA drugs and natural sleep and reveal a crucial role of the neuroendocrine system in regulating global brain states. VIDEO ABSTRACT.


Asunto(s)
Anestésicos Generales/farmacología , Hipnóticos y Sedantes/farmacología , Células Neuroendocrinas/efectos de los fármacos , Sueño de Onda Lenta/efectos de los fármacos , Núcleo Supraóptico/efectos de los fármacos , Anestesia General , Animales , Dexmedetomidina/farmacología , Electroencefalografía , Electromiografía , Fenómenos Electrofisiológicos , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Isoflurano/farmacología , Ketamina/farmacología , Ratones , Células Neuroendocrinas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Optogenética , Técnicas de Placa-Clamp , Propofol/farmacología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Sueño/efectos de los fármacos , Sueño/fisiología , Sueño de Onda Lenta/fisiología , Núcleo Supraóptico/citología , Núcleo Supraóptico/metabolismo
5.
Physiol Behav ; 196: 78-83, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30138634

RESUMEN

Although female rats are typically described as having a promiscuous mating strategy, if sexually naïve females have their formative sexually rewarding experiences paired with the same male, they will recognize that male and display mate-guarding behavior towards him in the presence of a female competitor. Female rats that display mate guarding behavior also show enhanced activation of oxytocin and vasopressin neurons in the supraoptic and paraventricular hypothalamic nucleus. Here, we examined the potential role that histone demethylation might have in establishing this pair-bonded behavior, and whether the corresponding changes in oxytocin and vasopressin neuronal activation depended on demethylation. To accomplish this, we examined the effect of a lysine-specific demethylase-1 inhibitor to block the action of demethylase enzymes and maintain the methylation state of corresponding genes. Female rats treated with the demethylase inhibitor failed to show any measure of mate guarding, whereas females treated with vehicle displayed mate guarding behavior. Demethylase inhibitor treatment also blocked the ability of familiar male cues to activate oxytocin and vasopressin neurons, whereas vehicle-treated females showed this enhanced activation. These data indicate that histone demethylation is a crucial component in the epigenetic modification of neural circuitry that underlies conditioned mate guarding in female rats. These results are the first to demonstrate the role of histone demethylation underlying changes in mating strategy.


Asunto(s)
Histona Demetilasas/metabolismo , Conducta Sexual Animal/fisiología , Animales , Conducta Competitiva/efectos de los fármacos , Conducta Competitiva/fisiología , Condicionamiento Psicológico/efectos de los fármacos , Condicionamiento Psicológico/fisiología , Femenino , Histona Demetilasas/antagonistas & inhibidores , Masculino , Oxitocina/metabolismo , Núcleo Hipotalámico Paraventricular/citología , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Ratas Long-Evans , Conducta Sexual Animal/efectos de los fármacos , Núcleo Supraóptico/citología , Núcleo Supraóptico/efectos de los fármacos , Núcleo Supraóptico/metabolismo , Vasopresinas/metabolismo
6.
J Neurophysiol ; 120(4): 1728-1739, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30020842

RESUMEN

Magnocellular oxytocin (OT) and vasopressin (VP) neurons express an afterhyperpolarization (AHP) following spike trains that attenuates firing rate and contributes to burst patterning. This AHP includes contributions from an apamin-sensitive, medium-duration AHP (mAHP) and from an apamin-insensitive, slow-duration AHP (sAHP). These AHPs are Ca2+ dependent and activated by Ca2+ influx through voltage-gated Ca2+ channels. Across central nervous system neurons that generate Ca2+-dependent AHPs, the Ca2+ channels that couple to the mAHP and sAHP differ greatly, but for magnocellular neurosecretory cells this relationship is unknown. Using simultaneous whole cell recording and Ca2+ imaging, we evaluated the effect of specific high-voltage-activated (HVA) Ca2+ channel blockers on the mAHP and sAHP. Block of all HVA channels via 400 µM Cd2+ inhibited almost the entire AHP. We tested nifedipine, conotoxin GVIA, agatoxin IVA, and SNX-482, specific blockers of L-, N-, P/Q-, and R-type channels, respectively. The N-type channel blocker conotoxin GVIA (1 µM) was the only toxin that inhibited the mAHP in either OT or VP neurons although the effect on VP neurons was weaker by comparison. The sAHP was significantly inhibited by N-type block in OT neurons and by R-type block in VP neurons although neither accounted for the entirety of the sAHP. Thus the mAHP appears to be elicited by Ca2+ from mostly N-type channels in both OT and VP neurons, but the contributions of specific Ca2+ channel types to the sAHP in each cell type are different. Alternative sources to HVA channels may contribute Ca2+ for the sAHP. NEW & NOTEWORTHY Despite the importance of afterhyperpolarization (AHP) mechanisms for regulating firing behavior of oxytocin (OT) and vasopressin (VP) neurons of supraoptic nucleus, which types of high-voltage-activated Ca2+ channels elicit AHPs in these cells was unknown. We found that N-type channels couple to the medium AHP in both cell types. For the slow AHP, N-type channels contribute in OT neurons, whereas R-type contribute in VP neurons. No single Ca2+ channel blocker abolished the entire AHP, suggesting that additional Ca2+ sources are involved.


Asunto(s)
Canales de Calcio Tipo N/metabolismo , Neuronas/metabolismo , Núcleo Supraóptico/metabolismo , Animales , Apamina/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Conotoxinas/farmacología , Femenino , Potenciales de la Membrana , Neuronas/efectos de los fármacos , Neuronas/fisiología , Nifedipino/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Ratas , Ratas Sprague-Dawley , Núcleo Supraóptico/citología , Núcleo Supraóptico/fisiología
7.
J Physiol ; 595(24): 7399-7411, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29034474

RESUMEN

KEY POINTS: NMDA receptor (NMDAR)-mediated Ca2+ signalling plays a critical role in modulating hypothalamic neurosecretory function. However, whether an altered NMDAR-evoked changes in Ca2+ (NMDAR-ΔCa2+ ) signalling in magnocellular neurosecretory cells (MNCs) may contribute to neurohumoral activation during disease states is unknown. We show that activation of NMDARs evoked similar inward currents in MNCs of sham and renovascular hypertensive (RVH) rats. Despite this, a prolonged and larger NMDAR-ΔCa2+ response was observed in the latter. The exacerbated NMDAR-ΔCa2+ responses in MNCs of RVH rats affected both somatic and dendritic compartments. Inhibition of the endoplasmic reticulum sarcoendoplasmic reticulum calcium trasport ATPase (SERCA) pump prolonged NMDAR-ΔCa2+ responses in sham rats, but not in RVH rats. Our study supports an altered spatiotemporal dynamic of NMDAR-ΔCa2+ signalling in MNCs from RVH rats, partly due to blunted endoplasmic reticulum Ca2+ buffering capacity. ABSTRACT: A growing body of evidence supports an elevated NMDA receptor (NMDAR)-mediated glutamate excitatory function in the supraoptic nucleus and paraventricular nucleus of hypertensive rats that contributes to neurohumoral activation in this disease. However, the precise mechanisms underlying altered NMDAR signalling in hypertension remain to be elucidated. In this study, we performed simultaneous electrophysiology and fast confocal Ca2+ imaging to determine whether altered NMDAR-mediated changes in intracellular Ca2+ levels (NMDAR-ΔCa2+ ) occurred in hypothalamic magnocellular neurosecretory cells (MNCs) in renovascular hypertensive (RVH) rats. We found that despite evoking a similar excitatory inward current, activation of NMDARs resulted in a larger and prolonged ΔCa2+ in MNCs from RVH rats. Changes in NMDAR-ΔCa2+ dynamics were observed both in somatic and dendritic compartments. Inhibition of the sarcoendoplasmic reticulum calcium trasport ATPase (SERCA) pump activity with thapsigargin prolonged NMDAR-ΔCa2+ responses in MNCs of sham rats, but this effect was occluded in RVH rats, thus equalizing the magnitude and time course of the NMDA-ΔCa2+ responses between the two experimental groups. Taken together, our results support (1) an exacerbated NMDAR-ΔCa2+ response in somatodendritic compartments of MNCs of RVH rats, and (2) that a blunted ER Ca2+ buffering capacity contributes to the altered NMDAR-ΔCa2+ dynamics in this condition. Thus, altered spatiotemporal dynamics of the NMDAR-ΔCa2+ response stands as an underlying mechanism contributing to neurohumoral activation in neurogenic hypertension.


Asunto(s)
Señalización del Calcio , Hipertensión/metabolismo , Neuronas/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Núcleo Supraóptico/metabolismo , Animales , Masculino , Núcleo Hipotalámico Paraventricular/citología , Ratas , Ratas Wistar , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Núcleo Supraóptico/citología
8.
PLoS One ; 12(7): e0180368, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28683135

RESUMEN

Integrate-and-fire (IF) models can provide close matches to the discharge activity of neurons, but do they oversimplify the biophysical properties of the neurons? A single compartment Hodgkin-Huxley (HH) model of the oxytocin neuron has previously been developed, incorporating biophysical measurements of channel properties obtained in vitro. A simpler modified integrate-and-fire model has also been developed, which can match well the characteristic spike patterning of oxytocin neurons as observed in vivo. Here, we extended the HH model to incorporate synaptic input, to enable us to compare spike activity in the model with experimental data obtained in vivo. We refined the HH model parameters to closely match the data, and then matched the same experimental data with a modified IF model, using an evolutionary algorithm to optimise parameter matching. Finally we compared the properties of the modified HH model with those of the IF model to seek an explanation for differences between spike patterning in vitro and in vivo. We show that, with slight modifications, the original HH model, like the IF model, is able to closely match both the interspike interval (ISI) distributions of oxytocin neurons and the observed variability of spike firing rates in vivo and in vitro. This close match of both models to data depends on the presence of a slow activity-dependent hyperpolarisation (AHP); this is represented in both models and the parameters used in the HH model representation match well with optimal parameters of the IF model found by an evolutionary algorithm. The ability of both models to fit data closely also depends on a shorter hyperpolarising after potential (HAP); this is explicitly represented in the IF model, but in the HH model, it emerges from a combination of several components. The critical elements of this combination are identified.


Asunto(s)
Potenciales de Acción/fisiología , Modelos Neurológicos , Neuronas/fisiología , Oxitocina/metabolismo , Núcleo Supraóptico/fisiología , Transmisión Sináptica/fisiología , Algoritmos , Animales , Simulación por Computador , Ácido Glutámico/metabolismo , Neuronas/citología , Neurotransmisores/metabolismo , Ratas , Programas Informáticos , Núcleo Supraóptico/citología , Sinapsis/fisiología , Ácido gamma-Aminobutírico/metabolismo
9.
Endocrinology ; 158(7): 2200-2211, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28430937

RESUMEN

Oxytocin is a potent anorexigen and is believed to have a role in satiety signaling. We developed rat models to study the activity of oxytocin neurons in response to voluntary consumption or oral gavage of foods using c-Fos immunohistochemistry and in vivo electrophysiology. Using c-Fos expression as an indirect marker of neural activation, we showed that the percentage of magnocellular oxytocin neurons expressing c-Fos increased with voluntary consumption of sweetened condensed milk (SCM). To model the effect of food in the stomach, we gavaged anesthetized rats with SCM. The percentage of supraoptic nucleus and paraventricular nucleus magnocellular oxytocin-immunoreactive neurons expressing c-Fos increased with SCM gavage but not with gastric distention. To further examine the activity of the supraoptic nucleus, we made in vivo electrophysiological recordings from SON neurons, where anesthetized rats were gavaged with SCM or single cream. Pharmacologically identified oxytocin neurons responded to SCM gavage with a linear, proportional, and sustained increase in firing rate, but cream gavage resulted in a transient reduction in firing rate. Blood glucose increased after SCM gavage but not cream gavage. Plasma osmolarity and plasma sodium were unchanged throughout. We show that in response to high-sugar, but not high-fat, food in the stomach, there is an increase in the activity of oxytocin neurons. This does not appear to be a consequence of stomach distention or changes in osmotic pressure. Our data suggest that the presence of specific foods with different macronutrient profiles in the stomach differentially regulates the activity of oxytocin neurons.


Asunto(s)
Carbohidratos de la Dieta/farmacología , Ingestión de Alimentos/fisiología , Neuronas/efectos de los fármacos , Núcleo Supraóptico/efectos de los fármacos , Animales , Dieta , Dieta Alta en Grasa , Fenómenos Electrofisiológicos/efectos de los fármacos , Inmunohistoquímica , Masculino , Neuronas/metabolismo , Oxitocina/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley , Núcleo Supraóptico/citología , Núcleo Supraóptico/fisiología
10.
Physiol Rep ; 5(8)2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28432255

RESUMEN

The magnocellular neurosecretory cells (MNCs) of the hypothalamus regulate body fluid balance by releasing the hormones vasopressin (VP) and oxytocin (OT) in an osmolality-dependent manner. Elevations of external osmolality increase MNC firing and hormone release. MNC osmosensitivity is largely due to activation of a mechanosensitive non-selective cation current that responds to osmotically-evoked changes in MNC volume and is mediated by an N-terminal variant of the TRPV1 channel (∆N TRPV1). We report a novel mechanism by which increases in osmolality may modulate ∆N TRPV1-mediated currents and thus influence MNC electrical behaviour. We showed previously that acute elevations of external osmolality activate the enzyme phospholipase C (PLC) in isolated MNCs. We now show that the osmotic activation of PLC has a time course and dose-dependence that is consistent with a role in MNC osmosensitivity and that it contributes to the osmotically-evoked increase in non-selective cation current in MNCs through a protein kinase C-dependent pathway. We furthermore show that the mechanism of osmotic activation of PLC requires an increase in internal Ca2+ that depends on influx through L-type Ca2+ channels. Our data therefore suggest that MNCs possess an osmotically-activated Ca2+-dependent PLC that contributes to the osmotic activation of ∆N TRPV1 and may therefore be important in MNC osmosensitivity and in central osmoregulation.


Asunto(s)
Potenciales de Acción , Calcio/metabolismo , Neuronas/metabolismo , Presión Osmótica , Núcleo Supraóptico/metabolismo , Canales Catiónicos TRPV/metabolismo , Fosfolipasas de Tipo C/metabolismo , Animales , Canales de Calcio Tipo L/metabolismo , Células Cultivadas , Masculino , Neuronas/fisiología , Ratas , Ratas Long-Evans , Núcleo Supraóptico/citología , Núcleo Supraóptico/fisiología
11.
Neuron ; 93(1): 57-65, 2017 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-27989461

RESUMEN

Ingestion of water and food are major hypo- and hyperosmotic challenges. To protect the body from osmotic stress, posterior pituitary-projecting, vasopressin-secreting neurons (VPpp neurons) counter osmotic perturbations by altering their release of vasopressin, which controls renal water excretion. Vasopressin levels begin to fall within minutes of water consumption, even prior to changes in blood osmolality. To ascertain the precise temporal dynamics by which water or food ingestion affect VPpp neuron activity, we directly recorded the spiking and calcium activity of genetically defined VPpp neurons. In states of elevated osmolality, water availability rapidly decreased VPpp neuron activity within seconds, beginning prior to water ingestion, upon presentation of water-predicting cues. In contrast, food availability following food restriction rapidly increased VPpp neuron activity within seconds, but only following feeding onset. These rapid and distinct changes in activity during drinking and feeding suggest diverse neural mechanisms underlying anticipatory regulation of VPpp neurons.


Asunto(s)
Arginina Vasopresina/metabolismo , Conducta de Ingestión de Líquido/fisiología , Conducta Alimentaria/fisiología , Neuronas/fisiología , Presión Osmótica , Animales , Señales (Psicología) , Ratones , Neuronas/metabolismo , Concentración Osmolar , Núcleo Supraóptico/citología , Vasopresinas/metabolismo
12.
Front Neural Circuits ; 10: 92, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27932956

RESUMEN

The arginine-vasopressin (AVP)-containing hypothalamic magnocellular neurosecretory neurons (VPMNNs) are known for their role in hydro-electrolytic balance control via their projections to the neurohypophysis. Recently, projections from these same neurons to hippocampus, habenula and other brain regions in which vasopressin infusion modulates contingent social and emotionally-affected behaviors, have been reported. Here, we present evidence that VPMNN collaterals also project to the amygdaloid complex, and establish synaptic connections with neurons in central amygdala (CeA). The density of AVP innervation in amygdala was substantially increased in adult rats that had experienced neonatal maternal separation (MS), consistent with our previous observations that MS enhances VPMNN number in the paraventricular (PVN) and supraoptic (SON) nuclei of the hypothalamus. In the CeA, V1a AVP receptor mRNA was only observed in GABAergic neurons, demonstrated by complete co-localization of V1a transcripts in neurons expressing Gad1 and Gad2 transcripts in CeA using the RNAscope method. V1b and V2 receptor mRNAs were not detected, using the same method. Water-deprivation (WD) for 24 h, which increased the metabolic activity of VPMNNs, also increased anxiety-like behavior measured using the elevated plus maze (EPM) test, and this effect was mimicked by bilateral microinfusion of AVP into the CeA. Anxious behavior induced by either WD or AVP infusion was reversed by CeA infusion of V1a antagonist. VPMNNs are thus a newly discovered source of CeA inhibitory circuit modulation, through which both early-life and adult stress coping signals are conveyed from the hypothalamus to the amygdala.


Asunto(s)
Antagonistas de los Receptores de Hormonas Antidiuréticas/farmacología , Ansiedad/metabolismo , Arginina Vasopresina/metabolismo , Núcleo Amigdalino Central , Glutamato Descarboxilasa/metabolismo , Hipotálamo , Neuronas , Receptores de Vasopresinas/metabolismo , Animales , Antagonistas de los Receptores de Hormonas Antidiuréticas/administración & dosificación , Ansiedad/inducido químicamente , Conducta Animal , Núcleo Amigdalino Central/citología , Núcleo Amigdalino Central/metabolismo , Modelos Animales de Enfermedad , Neuronas GABAérgicas/citología , Neuronas GABAérgicas/metabolismo , Hipotálamo/citología , Hipotálamo/metabolismo , Masculino , Privación Materna , Neuronas/citología , Neuronas/metabolismo , Núcleo Hipotalámico Paraventricular/citología , Núcleo Hipotalámico Paraventricular/metabolismo , Ratas Wistar , Núcleo Supraóptico/citología , Núcleo Supraóptico/metabolismo , Privación de Agua
13.
Neuron ; 89(6): 1291-1304, 2016 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-26948889

RESUMEN

Oxytocin (OT) is a neuropeptide elaborated by the hypothalamic paraventricular (PVN) and supraoptic (SON) nuclei. Magnocellular OT neurons of these nuclei innervate numerous forebrain regions and release OT into the blood from the posterior pituitary. The PVN also harbors parvocellular OT cells that project to the brainstem and spinal cord, but their function has not been directly assessed. Here, we identified a subset of approximately 30 parvocellular OT neurons, with collateral projections onto magnocellular OT neurons and neurons of deep layers of the spinal cord. Evoked OT release from these OT neurons suppresses nociception and promotes analgesia in an animal model of inflammatory pain. Our findings identify a new population of OT neurons that modulates nociception in a two tier process: (1) directly by release of OT from axons onto sensory spinal cord neurons and inhibiting their activity and (2) indirectly by stimulating OT release from SON neurons into the periphery.


Asunto(s)
Neuralgia/sangre , Neuralgia/fisiopatología , Neuronas/fisiología , Oxitocina/metabolismo , Núcleo Hipotalámico Paraventricular/citología , Núcleo Supraóptico/citología , Potenciales de Acción/efectos de los fármacos , Animales , Colecistoquinina/farmacología , Modelos Animales de Enfermedad , Antagonistas de Aminoácidos Excitadores/farmacología , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/fisiología , Inflamación/inducido químicamente , Inflamación/complicaciones , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/fisiología , Neuralgia/tratamiento farmacológico , Neuralgia/patología , Oxitocina/sangre , Oxitocina/genética , Quinoxalinas/farmacología , Ratas , Ratas Wistar , Receptores de Oxitocina/genética , Receptores de Oxitocina/metabolismo , Médula Espinal/citología , Transducción Genética , Vasopresinas/genética , Vasopresinas/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo
14.
J Neuroendocrinol ; 28(4)2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26813227

RESUMEN

In the ventral glial limitans (VGL) of the supraoptic nucleus (SON) of the rat, a unique astrocyte type is found with an ability to undergo striking morphological plasticity in response to a wide range of physiological stimulations such as chronic hypernatraemia. This includes a thinning of the VGL, which contains the somata and proximal processes of these astrocytes, as well as an almost complete withdrawal of their vertically-oriented distal processes. Currently, there is little information available on the types of astrocytes that reside in the SON-VGL and which of these exhibit state-dependent structural plasticity. To address this, we enabled the visualisation of single SON-VGL glia using two novel cell labelling techniques with fluorescence microscopy. First, we used an inducible genetic reporter mouse line that allowed the specific labelling of a low density of astrocytes expressing glutamate and aspartate transporter (GLAST)/excitatory amino acid transporter 1. This approach revealed a high degree of variability in the morphology of mouse SON-VGL astrocytes, in contrast to what has been reported for cortical astrocytes. Next, we used the DiOlistlic labelling approach to label single glial cells with DiI in the SON-VGL of rats. Astrocytes observed using this approach shared the morphological features of GLAST-expressing astrocytes in the mouse SON-VGL. Specific structural aspects of these cells were modified by chronic hypernatraemia achieved by 7-day salt loading. Notably, the average area of cells exhibiting protoplasmic features was significantly reduced in the horizontal plane, and the size of varicosities present on fibrous projections was significantly enlarged. These observations indicate that novel cell labelling methods can significantly advance our understanding of SON-VGL cells and reveal specific forms of morphological plasticity that can be driven by chronic hypernatraemia.


Asunto(s)
Astrocitos/citología , Astrocitos/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Cloruro de Sodio/farmacología , Núcleo Supraóptico/citología , Animales , Plasticidad de la Célula/efectos de los fármacos , Transportador 1 de Aminoácidos Excitadores/genética , Hipernatremia/patología , Masculino , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Ratas , Coloración y Etiquetado
15.
Horm Behav ; 79: 1-7, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26718226

RESUMEN

The neuropeptide oxytocin (OT) influences prosocial behavior(s), aggression, and stress responsiveness, and these diverse effects are regulated in a species- and context-specific manner. The naked mole-rat (Heterocephalus glaber) is a unique species with which to study context-dependent effects of OT, exhibiting a strict social hierarchy with behavioral specialization within the subordinate caste: soldiers are aggressive and defend colonies against unfamiliar conspecifics while workers are prosocial and contribute to in-colony behaviors such as pup care. To determine if OT is involved in subcaste-specific behaviors, we compared behavioral responses between workers and soldiers of both sexes during a modified resident/intruder paradigm, and quantified activation of OT neurons in the hypothalamic paraventricular nucleus (PVN) and supraoptic nucleus (SON) using the immediate-early-gene marker c-fos co-localized with OT neurons. Resident workers and soldiers were age-matched with unfamiliar worker stimulus animals as intruders, and encounters were videorecorded and scored for aggressive behaviors. Colony-matched controls were left in their home colony for the duration of the encounters. Brains were extracted and cell counts were conducted for OT immunoreactive (ir), c-fos-ir, and percentage of OT-c-fos double-labeled cells. Results indicate that resident workers were less aggressive but showed greater OT neural activity than soldiers. Furthermore, a linear model including social treatment, cortisol, and subcaste revealed that subcaste was the only significant predictor of OT-c-fos double-labeled cells in the PVN. These data suggest that in naked mole-rats OT promotes prosocial behaviors rather than aggression and that even within subordinates status exerts robust effects on brain and behavior.


Asunto(s)
Conducta Animal/efectos de los fármacos , Jerarquia Social , Neuronas/efectos de los fármacos , Oxitocina/farmacología , Conducta Social , Agresión/efectos de los fármacos , Animales , Conducta Animal/fisiología , Encéfalo/efectos de los fármacos , Femenino , Masculino , Ratas Topo , Neuronas/fisiología , Núcleo Hipotalámico Paraventricular/citología , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Supraóptico/citología , Núcleo Supraóptico/efectos de los fármacos
16.
Artículo en Inglés | MEDLINE | ID: mdl-28056741

RESUMEN

BACKGROUND: Magnocellular neurosecretory neurons of the hypothalamic supraoptic (SON) and paraventricular (PVN) nuclei synthesize vasopressin and oxytocin in response to signals generated by osmoreceptors and baroreceptors and, respectively, by receptors of the nipples and cervix. METHODS: We analyzed the literature identifying relevant articles dealing with synaptic inputs of neural afferent pathways to Vasopressin-and Oxytocin-secreting neurons of SON and PVN. RESULTS: This article focuses on the multisynaptic pathways involved in the regulation of Vasopressin and Oxytocin secretion. CONCLUSION: An updated topographic description of the afferent pathways involved in the regulation of VPergic and OTergic neurons and their synaptic inputs inducing the stimulus-secretion-coupling has been depicted.


Asunto(s)
Vías Aferentes/fisiología , Neuronas Aferentes/fisiología , Oxitocina/metabolismo , Núcleo Hipotalámico Paraventricular/citología , Núcleo Supraóptico/citología , Vasopresinas/metabolismo , Animales , Humanos , Neuronas Aferentes/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Hipotalámico Paraventricular/fisiología , Núcleo Supraóptico/metabolismo , Núcleo Supraóptico/fisiología , Transmisión Sináptica/fisiología
17.
J Neuroendocrinol ; 28(4)2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26610724

RESUMEN

Central administration of neurokinin B (NKB) agonists stimulates immediate early gene expression in the hypothalamus and increases the secretion of vasopressin from the posterior pituitary through a mechanism that depends on the activation of neurokinin receptor 3 receptors (NK3R). The present study reports that, in the rat, immunoreactivity for NK3R is expressed in magnocellular vasopressin and oxytocin neurones in the supraoptic nucleus (SON) and paraventricular nucleus (PVN) of the hypothalamus, and that NKB immunoreactivity is expressed in fibres in close juxtaposition with vasopressin neurones at both of these sites. Retrograde tracing in the rat shows that some NKB-expressing neurones in the arcuate nucleus project to the SON and, in mice, using an anterograde tracing approach, it is found that kisspeptin-expressing neurones of the arcuate nucleus, which are known to co-express NKB, project to the SON and PVN. Finally, i.c.v. injection of the NK3R agonist senktide is shown to potently increase the electrical activity of vasopressin neurones in the SON in vivo with no significant effect detected on oxytocin neurones. The results suggest that NKB-containing neurones in the arcuate nucleus regulate the secretion of vasopressin from magnocellular neurones in rodents, and the possible significance of this is discussed.


Asunto(s)
Núcleo Arqueado del Hipotálamo/fisiología , Neuroquinina B/metabolismo , Neuronas/fisiología , Núcleo Supraóptico/citología , Vasopresinas/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Infusiones Intraventriculares , Kisspeptinas/genética , Kisspeptinas/metabolismo , Masculino , Ratones , Ratones Transgénicos , Técnicas de Trazados de Vías Neuroanatómicas , Neuronas/metabolismo , Oxitocina/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/farmacología , Ratas , Receptores de Neuroquinina-3/agonistas , Receptores de Neuroquinina-3/metabolismo , Sustancia P/administración & dosificación , Sustancia P/análogos & derivados , Sustancia P/farmacología , Núcleo Supraóptico/metabolismo
18.
J Neuroendocrinol ; 28(4)2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26715365

RESUMEN

Oxytocin neurones of the rat supraoptic nucleus are osmoresponsive and, with all other things being equal, they fire at a mean rate that is proportional to the plasma sodium concentration. However, individual spike times are governed by highly stochastic events, namely the random occurrences of excitatory synaptic inputs, the probability of which is increased by increasing extracellular osmotic pressure. Accordingly, interspike intervals (ISIs) are very irregular. In the present study, we show, by statistical analyses of firing patterns in oxytocin neurones, that the mean firing rate as measured in bins of a few seconds is more regular than expected from the variability of ISIs. This is consistent with an intrinsic activity-dependent negative-feedback mechanism. To test this, we compared observed neuronal firing patterns with firing patterns generated by a leaky integrate-and-fire model neurone, modified to exhibit activity-dependent mechanisms known to be present in oxytocin neurones. The presence of a prolonged afterhyperpolarisation (AHP) was critical for the ability to mimic the observed regularisation of mean firing rate, although we also had to add a depolarising afterpotential (DAP; sometimes called an afterdepolarisation) to the model to match the observed ISI distributions. We tested this model by comparing its behaviour with the behaviour of oxytocin neurones exposed to apamin, a blocker of the medium AHP. Good fits indicate that the medium AHP actively contributes to the firing patterns of oxytocin neurones during non-bursting activity, and that oxytocin neurones generally express a DAP, even though this is usually masked by superposition of a larger AHP.


Asunto(s)
Potenciales de Acción/fisiología , Neuronas/fisiología , Oxitocina/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Apamina/farmacología , Modelos Neurológicos , Neuronas/efectos de los fármacos , Presión Osmótica/fisiología , Ratas , Núcleo Supraóptico/citología , Núcleo Supraóptico/efectos de los fármacos , Núcleo Supraóptico/fisiología
19.
Sheng Li Xue Bao ; 67(6): 583-90, 2015 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-26701633

RESUMEN

The present study was designed to investigate the inhibitory effects of intravenous general anesthetic propofol (0.1-3.0 mmol/L) on excitatory synaptic transmission in supraoptic nucleus (SON) neurons of rats, and to explore the underlying mechanisms by using intracellular recording technique and hypothalamic slice preparation. It was observed that stimulation of the dorsolateral region of SON could elicit the postsynaptic potentials (PSPs) in SON neurons. Of the 8 tested SON neurons, the PSPs of 7 (88%, 7/8) neurons were decreased by propofol in a concentration-dependent manner, in terms of the PSPs' amplitude (P < 0.01), area under curve, duration, half-width and 10%-90% decay time (P < 0.05). The PSPs were completely and reversibly abolished by 1.0 mmol/L propofol at 2 out of 7 tested cells. The depolarization responses induced by pressure ejection of exogenous glutamate were reversibly and concentration-dependently decreased by bath application of propofol. The PSPs and glutamate-induced responses recorded simultaneously were reversibly and concentration-dependently decreased by propofol, but 0.3 mmol/L propofol only abolished PSPs. The excitatory postsynaptic potentials (EPSPs) of 7 cells increased in the condition of picrotoxin (30 µmol/L, a GABA(A) receptor antagonist) pretreatment. On this basis, the inhibitory effects of propofol on EPSPs were decreased. These data indicate that the presynaptic and postsynaptic mechanisms may be both involved in the inhibitory effects of propofol on excitatory synaptic transmission in SON neurons. The inhibitory effects of propofol on excitatory synaptic transmission of SON neurons may be related to the activation of GABA(A) receptors, but at a high concentration, propofol may also act directly on glutamate receptors.


Asunto(s)
Potenciales Postsinápticos Excitadores/efectos de los fármacos , Neuronas/efectos de los fármacos , Propofol/farmacología , Núcleo Supraóptico/citología , Anestésicos Intravenosos/farmacología , Animales , Antagonistas de Receptores de GABA-A/farmacología , Ácido Glutámico/farmacología , Técnicas In Vitro , Ratas , Receptores de Glutamato/metabolismo
20.
J Neuroendocrinol ; 27(11): 835-49, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26403275

RESUMEN

An accumulating body of evidence suggests that the activity of the mineralocorticoid, aldosterone, in the brain via the mineralocorticoid receptor (MR) plays an important role in the regulation of blood pressure. MR was recently found in vasopressin and oxytocin synthesising magnocellular neurosecretory cells (MNCs) in both the paraventricular (PVN) and supraoptic (SON) nuclei in the hypothalamus. Considering the physiological effects of these hormones, MR in these neurones may be an important site mediating the action of aldosterone in blood pressure regulation within the brain. However, aldosterone activation of MR in the hypothalamus remains controversial as a result of the high binding affinity of glucocorticoids to MR at substantially higher concentrations compared to aldosterone. In aldosterone-sensitive epithelia, the enzyme 11ß-hydroxysteroid dehydrogenase type 2 (11ß-HSD2) prevents glucocorticoids from binding to MR by converting glucocorticoids into inactive metabolites. The present study aimed to determine whether 11ß-HSD2, which increases aldosterone selectivity, is expressed in MNCs. Specific 11ß-HSD2 immunoreactivity was found in the cytoplasm of the MNCs in both the SON and PVN. In addition, double-fluorescence confocal microscopy demonstrated that MR-immunoreactivity and 11ß-HSD2-in situ hybridised products are colocalised in MNCs. Lastly, single-cell reverse transcriptase-polymerase chain reaction detected MR and 11ß-HSD2 mRNAs from cDNA libraries derived from single identified MNCs. These findings strongly suggest that MNCs in the SON and PVN are aldosterone-sensitive neurones.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Sistemas Neurosecretores/citología , Sistemas Neurosecretores/metabolismo , Núcleo Hipotalámico Paraventricular/citología , Receptores de Mineralocorticoides/metabolismo , Núcleo Supraóptico/citología , Animales , Masculino , Sistemas Neurosecretores/enzimología , Núcleo Hipotalámico Paraventricular/enzimología , Núcleo Hipotalámico Paraventricular/metabolismo , Ratas , Núcleo Supraóptico/enzimología , Núcleo Supraóptico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...