Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 625(7996): 743-749, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38233522

RESUMEN

Survival requires the selection of appropriate behaviour in response to threats, and dysregulated defensive reactions are associated with psychiatric illnesses such as post-traumatic stress and panic disorder1. Threat-induced behaviours, including freezing and flight, are controlled by neuronal circuits in the central amygdala (CeA)2; however, the source of neuronal excitation of the CeA that contributes to high-intensity defensive responses is unknown. Here we used a combination of neuroanatomical mapping, in vivo calcium imaging, functional manipulations and electrophysiology to characterize a previously unknown projection from the dorsal peduncular (DP) prefrontal cortex to the CeA. DP-to-CeA neurons are glutamatergic and specifically target the medial CeA, the main amygdalar output nucleus mediating conditioned responses to threat. Using a behavioural paradigm that elicits both conditioned freezing and flight, we found that CeA-projecting DP neurons are activated by high-intensity threats in a context-dependent manner. Functional manipulations revealed that the DP-to-CeA pathway is necessary and sufficient for both avoidance behaviour and flight. Furthermore, we found that DP neurons synapse onto neurons within the medial CeA that project to midbrain flight centres. These results elucidate a non-canonical top-down pathway regulating defensive responses.


Asunto(s)
Reacción de Prevención , Núcleo Amigdalino Central , Vías Nerviosas , Neuronas , Reacción de Prevención/fisiología , Núcleo Amigdalino Central/citología , Núcleo Amigdalino Central/fisiología , Neuronas/fisiología , Corteza Prefrontal/citología , Corteza Prefrontal/fisiología , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Ácido Glutámico/metabolismo , Vías Nerviosas/fisiología , Calcio/análisis , Electrofisiología , Puente/citología , Puente/fisiología
2.
J Neurosci ; 43(47): 7902-7912, 2023 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-37739795

RESUMEN

Chronic alcohol exposure leads to a neuroinflammatory response involving activation of the nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome and proinflammatory cytokine production. Acute ethanol (EtOH) exposure activates GABAergic synapses in the central and basolateral amygdala (BLA) ex vivo, but whether this rapid modulation of synaptic inhibition is because of an acute inflammatory response and alters anxiety-like behavior in male and female animals is not known. Here, we tested the hypotheses that acute EtOH facilitates inhibitory synaptic transmission in the BLA by activating the NLRP3 inflammasome-dependent acute inflammatory response, that the alcohol-induced increase in inhibition is cell type and sex dependent, and that acute EtOH in the BLA reduces anxiety-like behavior. Acute EtOH application at a binge-like concentration (22-44 mm) stimulated synaptic GABA release from putative parvalbumin (PV) interneurons onto BLA principal neurons in ex vivo brain slices from male, but not female, rats. The EtOH facilitation of synaptic inhibition was blocked by antagonists of the Toll-like receptor 4 (TLR4), the NLRP3 inflammasome, and interleukin-1 receptors, suggesting it was mediated by a rapid local neuroinflammatory response in the BLA. In vivo, bilateral injection of EtOH directly into the BLA produced an acute concentration-dependent reduction in anxiety-like behavior in male but not female rats. These findings demonstrate that acute EtOH in the BLA regulates anxiety-like behavior in a sex-dependent manner and suggest that this effect is associated with presynaptic facilitation of parvalbumin-expressing interneuron inputs to BLA principal neurons via a local NLRP3 inflammasome-dependent neuroimmune response.SIGNIFICANCE STATEMENT Chronic alcohol exposure produces a neuroinflammatory response, which contributes to alcohol-associated pathologies. Acute alcohol administration increases inhibitory synaptic signaling in the brain, but the mechanism for the rapid alcohol facilitation of inhibitory circuits is unknown. We found that acute ethanol at binge-like concentrations in the basolateral amygdala (BLA) facilitates GABA release from parvalbumin-expressing (PV) interneuron synapses onto principal neurons in ex vivo brain slices from male rats and that intra-BLA ethanol reduces anxiety-like behavior in vivo in male rats, but not female rats. The ethanol (EtOH) facilitation of inhibition in the BLA is mediated by Toll-like receptor 4 (TLR4) and nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome activation and proinflammatory IL-1ß signaling, which suggests a rapid NLRP3 inflammasome-dependent neuroimmune cascade that plays a critical role in acute alcohol intoxication.


Asunto(s)
Ansiedad , Complejo Nuclear Basolateral , Etanol , Animales , Femenino , Masculino , Ratas , Ansiedad/inducido químicamente , Ansiedad/metabolismo , Complejo Nuclear Basolateral/efectos de los fármacos , Complejo Nuclear Basolateral/metabolismo , Etanol/toxicidad , Ácido gamma-Aminobutírico/metabolismo , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Nucleótidos/metabolismo , Nucleótidos/farmacología , Parvalbúminas/metabolismo , Receptor Toll-Like 4/metabolismo
3.
J Neurosci ; 36(32): 8461-70, 2016 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-27511017

RESUMEN

UNLABELLED: Stress and glucocorticoids stimulate the rapid mobilization of endocannabinoids in the basolateral amygdala (BLA). Cannabinoid receptors in the BLA contribute to anxiogenesis and fear-memory formation. We tested for rapid glucocorticoid-induced endocannabinoid regulation of synaptic inhibition in the rat BLA. Glucocorticoid application to amygdala slices elicited a rapid, nonreversible suppression of spontaneous, but not evoked, GABAergic synaptic currents in BLA principal neurons; the effect was also seen with a membrane-impermeant glucocorticoid, but not with intracellular glucocorticoid application, implicating a membrane-associated glucocorticoid receptor. The glucocorticoid suppression of GABA currents was not blocked by antagonists of nuclear corticosteroid receptors, or by inhibitors of gene transcription or protein synthesis, but was blocked by inhibiting postsynaptic G-protein activity, suggesting a postsynaptic nongenomic steroid signaling mechanism that stimulates the release of a retrograde messenger. The rapid glucocorticoid-induced suppression of inhibition was prevented by blocking CB1 receptors and 2-arachidonoylglycerol (2-AG) synthesis, and it was mimicked and occluded by CB1 receptor agonists, indicating it was mediated by the retrograde release of the endocannabinoid 2-AG. The rapid glucocorticoid effect in BLA neurons in vitro was occluded by prior in vivo acute stress-induced, or prior in vitro glucocorticoid-induced, release of endocannabinoid. Acute stress also caused an increase in anxiety-like behavior that was attenuated by blocking CB1 receptor activation and inhibiting 2-AG synthesis in the BLA. Together, these findings suggest that acute stress causes a long-lasting suppression of synaptic inhibition in BLA neurons via a membrane glucocorticoid receptor-induced release of 2-AG at GABA synapses, which contributes to stress-induced anxiogenesis. SIGNIFICANCE STATEMENT: We provide a cellular mechanism in the basolateral amygdala (BLA) for the rapid stress regulation of anxiogenesis in rats. We demonstrate a nongenomic glucocorticoid induction of long-lasting suppression of synaptic inhibition that is mediated by retrograde endocannabinoid release at GABA synapses. The rapid glucocorticoid-induced endocannabinoid suppression of synaptic inhibition is initiated by a membrane-associated glucocorticoid receptor in BLA principal neurons. We show that acute stress increases anxiety-like behavior via an endocannabinoid-dependent mechanism centered in the BLA. The stress-induced endocannabinoid modulation of synaptic transmission in the BLA contributes, therefore, to the stress regulation of anxiety, and may play a role in anxiety disorders of the amygdala.


Asunto(s)
Ansiedad/patología , Complejo Nuclear Basolateral/metabolismo , Endocannabinoides/metabolismo , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Restricción Física/fisiología , Animales , Antieméticos/farmacología , Ansiedad/fisiopatología , Ácidos Araquidónicos/farmacología , Complejo Nuclear Basolateral/efectos de los fármacos , Benzoxazinas/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Agonistas de Receptores de Cannabinoides/farmacología , Antagonistas de Receptores de Cannabinoides/farmacología , Dexametasona/farmacología , Modelos Animales de Enfermedad , Endocannabinoides/farmacología , Inhibidores Enzimáticos/farmacología , Glucocorticoides/farmacología , Glicéridos/farmacología , Masculino , Morfolinas/farmacología , Naftalenos/farmacología , Piperidinas/farmacología , Pirazoles/farmacología , Ratas , Ratas Wistar , Rimonabant , Transmisión Sináptica
4.
J Neurosci ; 34(18): 6201-13, 2014 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-24790191

RESUMEN

Behavioral and physiological coupling between energy balance and fluid homeostasis is critical for survival. The orexigenic hormone ghrelin has been shown to stimulate the secretion of the osmoregulatory hormone vasopressin (VP), linking nutritional status to the control of blood osmolality, although the mechanism of this systemic crosstalk is unknown. Here, we show using electrophysiological recordings and calcium imaging in rat brain slices that ghrelin stimulates VP neurons in the hypothalamic paraventricular nucleus (PVN) in a nutritional state-dependent manner by activating an excitatory GABAergic synaptic input via a retrograde neuronal-glial circuit. In slices from fasted rats, ghrelin activation of a postsynaptic ghrelin receptor, the growth hormone secretagogue receptor type 1a (GHS-R1a), in VP neurons caused the dendritic release of VP, which stimulated astrocytes to release the gliotransmitter adenosine triphosphate (ATP). ATP activation of P2X receptors excited presynaptic GABA neurons to increase GABA release, which was excitatory to the VP neurons. This trans-neuronal-glial retrograde circuit activated by ghrelin provides an alternative means of stimulation of VP release and represents a novel mechanism of neuronal control by local neuronal-glial circuits. It also provides a potential cellular mechanism for the physiological integration of energy and fluid homeostasis.


Asunto(s)
Ghrelina/farmacología , Neuroglía/metabolismo , Neuronas/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/citología , Vasopresinas/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Adenosina Trifosfato/metabolismo , Anestésicos Locales/farmacología , Animales , Calcio/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Técnicas In Vitro , Masculino , Red Nerviosa/efectos de los fármacos , Red Nerviosa/fisiología , Neuroglía/efectos de los fármacos , Neuronas/fisiología , Neurotransmisores/farmacología , Ratas , Ratas Wistar , Potenciales Sinápticos/efectos de los fármacos , Potenciales Sinápticos/genética , Tetrodotoxina/farmacología , Vasopresinas/genética
5.
Front Neuroendocrinol ; 35(1): 72-5, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24103541

RESUMEN

Glucocorticoids (GCs) are a class of steroid hormones that have been known to be involved in various physiological processes and to play a pivotal role in preserving basal and stress-related homeostasis. GCs are also widely used clinically as anti-inflammatory, immunosuppressive, anti-shock drugs. It is believed traditionally that GCs exert most of their effects genomically. In addition to the well-known classical genomic mechanisms, GCs also affect various functions via rapid, nongenomic mechanisms. The therapeutic benefits of nongenomic GC actions have been exploited in clinical medicine, especially with high-dose pulsed glucocorticoid administration. However, it is certainly not the case that the inherent nongenomic glucocorticoid mechanisms evolved only for their clinical utility. Here, we review the recent literature on nongenomic actions of GCs related to stress and the physiological significance of these actions, and we propose reasons why nongenomic mechanisms of GC actions are needed.


Asunto(s)
Antiinflamatorios/farmacología , Glucocorticoides/metabolismo , Inflamación/tratamiento farmacológico , Estrés Fisiológico/efectos de los fármacos , Animales , Humanos , Inflamación/metabolismo , Receptores de Glucocorticoides/metabolismo , Esteroides/metabolismo
6.
J Neurosci ; 33(46): 18331-42, 2013 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-24227742

RESUMEN

Cannabinoid receptors are functionally operant at both glutamate and GABA synapses on hypothalamic magnocellular neuroendocrine cells; however, retrograde endocannabinoid actions are evoked at only glutamate synapses. We tested whether the functional targeting of evoked retrograde endocannabinoid actions to glutamate, and not GABA, synapses on magnocellular neurons is the result of the spatial restriction of extracellular endocannabinoids by astrocytes. Whole-cell GABA synaptic currents were recorded in magnocellular neurons in rat hypothalamic slices following manipulations to reduce glial buffering of extracellular signals. Depolarization- and glucocorticoid-evoked retrograde endocannabinoid suppression of synaptic GABA release was not detected under normal conditions, but occurred in both oxytocin and vasopressin neurons under conditions of attenuated glial coverage and depressed glial metabolic function, suggesting an emergent endocannabinoid modulation of GABA synapses with the loss of astrocyte function. Tonic endocannabinoid suppression of GABA release was insensitive to glial manipulation. Blocking cannabinoid transport mimicked, and increasing the extracellular viscosity reversed, the effect of suppressed glial buffering on the endocannabinoid modulation of GABA release. Evoked, but not tonic, endocannabinoid modulation of GABA synapses was mediated by 2-arachidonoylglycerol. Therefore, depolarization- and glucocorticoid-evoked 2-arachidonoylglycerol release from magnocellular neurons is spatially restricted to glutamate synapses by astrocytes, but spills over onto GABA synapses under conditions of reduced astrocyte buffering; tonic endocannabinoid modulation of GABA release, in contrast, is likely mediated by anandamide and is insensitive to astrocytic buffering. Astrocytes, therefore, provide dynamic control of stimulus-evoked 2-arachidonoylglycerol, but not tonic anandamide, regulation of GABA synaptic inputs to magnocellular neuroendocrine cells under different physiological conditions.


Asunto(s)
Endocannabinoides/fisiología , Hipotálamo/fisiología , Células Neuroendocrinas/fisiología , Neuroglía/fisiología , Sinapsis/fisiología , Animales , Masculino , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley
7.
J Physiol ; 592(19): 4221-33, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25063825

RESUMEN

The magnocellular vasopressin (VP) and oxytocin (OT) neurones undergo long-term synaptic plasticity to accommodate prolonged hormone demand. By contrast, rapidly induced,transient synaptic plasticity in response to brief stimuli could enable the activation of magnocellular neurones in response to acute challenges. Here, we report a robust short-term potentiation of asynchronous GABAergic synaptic inputs (STP(GABA)) to VP and OT neurones of the hypothalamic supraoptic nucleus elicited by repetitive extracellular electrical stimulation.The STP(GABA) required extracellular Ca2+, but did not require activation of glutamate, VP or OT receptors or nitric oxide synthesis. Presynaptic action potential generation was necessary for the induction, but not the maintenance, of STP(GABA). The STP(GABA) led to a minutes-long GABA(A)receptor-dependent increase in spike frequency in VP neurones, but not in OT neurones,consistent with an excitatory function of GABA in only VP neurones and with the generation of prolonged bursts of action potentials in VP neurones. Therefore, this short-term plasticity of GABAergic synaptic inputs is likely to play very different roles in the regulation of OT and VP neurones and their distinct patterns of physiological activation.


Asunto(s)
Neuronas GABAérgicas/fisiología , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Oxitocina/metabolismo , Núcleo Supraóptico/fisiología , Sinapsis/metabolismo , Vasopresinas/metabolismo , Potenciales de Acción/fisiología , Animales , Estimulación Eléctrica , Neuronas GABAérgicas/citología , Neuronas GABAérgicas/metabolismo , Neuronas/metabolismo , Ratas , Ratas Wistar , Núcleo Supraóptico/citología , Núcleo Supraóptico/metabolismo
8.
J Neurosci ; 32(2): 572-82, 2012 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-22238092

RESUMEN

Neuronal excitability in the adult brain is controlled by a balance between synaptic excitation and inhibition mediated by glutamate and GABA, respectively. While generally inhibitory in the adult brain, GABA(A) receptor activation is excitatory under certain conditions in which the GABA reversal potential is shifted positive due to intracellular Cl(-) accumulation, such as during early postnatal development and brain injury. However, the conditions under which GABA is excitatory are generally either transitory or pathological. Here, we reveal GABAergic synaptic inputs to be uniformly excitatory in vasopressin (VP)-secreting magnocellular neurons in the adult hypothalamus under normal conditions. The GABA reversal potential (E(GABA)) was positive to resting potential and spike threshold in VP neurons, but not in oxytocin (OT)-secreting neurons. The VP neurons lacked expression of the K(+)-Cl(-) cotransporter 2 (KCC2), the predominant Cl(-) exporter in the adult brain. The E(GABA) was unaffected by inhibition of KCC2 in VP neurons, but was shifted positive in OT neurons, which express KCC2. Alternatively, inhibition of the Na(+)-K(+)-Cl(-) cotransporter 1 (NKCC1), a Cl(-) importer expressed in most cell types mainly during postnatal development, caused a negative shift in E(GABA) in VP neurons, but had no effect on GABA currents in OT neurons. GABA(A) receptor blockade caused a decrease in the firing rate of VP neurons, but an increase in firing in OT neurons. Our findings demonstrate that GABA is excitatory in adult VP neurons, suggesting that the classical excitation/inhibition paradigm of synaptic glutamate and GABA control of neuronal excitability does not apply to VP neurons.


Asunto(s)
Potenciales Postsinápticos Excitadores/fisiología , Células Neuroendocrinas/fisiología , Núcleo Hipotalámico Paraventricular/fisiología , Núcleo Supraóptico/fisiología , Vasopresinas/fisiología , Ácido gamma-Aminobutírico/fisiología , Envejecimiento/fisiología , Animales , Neuronas GABAérgicas/fisiología , Masculino , Núcleo Hipotalámico Paraventricular/citología , Ratas , Ratas Transgénicas , Ratas Wistar , Núcleo Supraóptico/citología
9.
Endocrinology ; 164(11)2023 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-37788632

RESUMEN

Stress induces changes in nervous system function on different signaling levels, from molecular signaling to synaptic transmission to neural circuits to behavior-and on different time scales, from rapid onset and transient to delayed and long-lasting. The principal effectors of stress plasticity are glucocorticoids, steroid hormones that act with a broad range of signaling competency due to the expression of multiple nuclear and membrane receptor subtypes in virtually every tissue of the organism. Glucocorticoid and mineralocorticoid receptors are localized to each of the cellular compartments of the receptor-expressing cells-the membrane, cytosol, and nucleus. In this review, we cover the neuroendocrine effects of stress, focusing mainly on the rapid actions of acute stress-induced glucocorticoids that effect changes in synaptic transmission and neuronal excitability by modulating synaptic and intrinsic neuronal properties via activation of presumed membrane glucocorticoid and mineralocorticoid receptors. We describe the synaptic plasticity that occurs in 4 stress-associated brain structures, the hypothalamus, hippocampus, amygdala, and prefrontal cortex, in response to single or short-term stress exposure. The rapid transformative impact of glucocorticoids makes this stress signal a particularly potent effector of acute neuronal plasticity.


Asunto(s)
Glucocorticoides , Receptores de Mineralocorticoides , Glucocorticoides/farmacología , Plasticidad Neuronal , Encéfalo , Transmisión Sináptica , Receptores de Glucocorticoides/fisiología , Estrés Psicológico
10.
eNeuro ; 10(9)2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37640541

RESUMEN

To survive, animals must meet their biological needs while simultaneously avoiding danger. However, the neurobiological basis of appetitive and aversive survival behaviors has historically been studied using separate behavioral tasks. While recent studies in mice have quantified appetitive and aversive conditioned responses simultaneously (Jikomes et al., 2016; Heinz et al., 2017), these tasks required different behavioral responses to each stimulus. As many brain regions involved in survival behavior process stimuli of opposite valence, we developed a paradigm in which mice perform the same response (nose poke) to distinct auditory cues to obtain a rewarding outcome (palatable food) or avoid an aversive outcome (mild footshoock). This design allows for both within-subject and between-subject comparisons as animals respond to appetitive and aversive cues. The central nucleus of the amygdala (CeA) is implicated in the regulation of responses to stimuli of either valence. Considering its role in threat processing (Wilensky et al., 2006; Haubensak et al., 2010) and regulation of incentive salience (Warlow and Berridge, 2021), it is important to examine the contribution of the CeA to mechanisms potentially underlying comorbid dysregulation of avoidance and reward (Sinha, 2008; Bolton et al., 2009). Using this paradigm, we tested the role of two molecularly defined CeA subtypes previously linked to consummatory and defensive behaviors. Significant strain differences in the acquisition and performance of the task were observed. Bidirectional chemogenetic manipulation of CeA somatostatin (SOM) neurons altered motivation for reward and perseveration of reward-seeking responses on avoidance trials. Manipulation of corticotropin-releasing factor neurons (CRF) had no significant effect on food reward consumption, motivation, or task performance. This paradigm will facilitate investigations into the neuronal mechanisms controlling motivated behavior across valences.


Asunto(s)
Núcleo Amigdalino Central , Animales , Ratones , Condicionamiento Operante , Motivación , Afecto , Neuronas
11.
bioRxiv ; 2023 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-37461627

RESUMEN

To survive, animals must meet their biological needs while simultaneously avoiding danger. However, the neurobiological basis of appetitive and aversive survival behaviors has historically been studied using separate behavioral tasks. While recent studies in mice have quantified appetitive and aversive conditioned responses simultaneously (Heinz et al., 2017; Jikomes et al., 2016), these tasks required different behavioral responses to each stimulus. As many brain regions involved in survival behavior process stimuli of opposite valence, we developed a paradigm in which mice perform the same response (nosepoke) to distinct auditory cues to obtain a rewarding outcome (palatable food) or avoid an aversive outcome (mild footshoock). This design allows for both within- and between-subject comparisons as animals respond to appetitive and aversive cues. The central nucleus of the amygdala (CeA) is implicated in the regulation of responses to stimuli of either valence. Considering its role in threat processing (Haubensak et al., 2010; Wilensky et al., 2006) and regulation of incentive salience (Warlow and Berridge, 2021), it is important to examine the contribution of the CeA to mechanisms potentially underlying comorbid dysregulation of avoidance and reward (Bolton et al., 2009; Sinha, 2008). Using this paradigm, we tested the role of two molecularly defined CeA subtypes previously linked to consummatory and defensive behaviors. Significant strain differences in the acquisition and performance of the task were observed. Bidirectional chemogenetic manipulation of CeA somatostatin (SOM) neurons altered motivation for reward and perseveration of reward-seeking responses on avoidance trials. Manipulation of corticotropin-releasing factor neurons (CRF) had no significant effect on food reward consumption, motivation, or task performance. This paradigm will facilitate investigations into the neuronal mechanisms controlling motivated behavior across valences. Significance Statement: It is unclear how different neuronal populations contribute to reward- and aversion-driven behaviors within a subject. To address this question, we developed a novel behavioral paradigm in which mice obtain food and avoid footshocks via the same operant response. We then use this paradigm to test how the central amygdala coordinates appetitive and aversive behavioral responses. By testing somatostatin-IRES-Cre and CRF-IRES-Cre transgenic lines, we found significant differences between strains on task acquisition and performance. Using chemogenetics, we demonstrate that CeA SOM+ neurons regulate motivation for reward, while manipulation of CeA CRF+ neurons had no effect on task performance. Future studies investigating the interaction between positive and negative motivation circuits should benefit from the use of this dual valence paradigm.

12.
Network ; 23(1-2): 59-75, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22571251

RESUMEN

The coupled oscillator model has previously been used for the simulation of neuronal activities in in vitro rat hippocampal slice seizure data and the evaluation of seizure suppression algorithms. Each model unit can be described as either an oscillator which can generate action potential spike trains without inputs, or a threshold-based unit. With the change of only one parameter, each unit can either be an oscillator or a threshold-based spiking unit. This would eliminate the need of a new set of equations for each type of unit. Previous analysis has suggested that long kernel duration and imbalance of inhibitory feedback can cause the system to intermittently transition into and out of ictal activities. The state transitions of seizure-like events were investigated here; specifically, how the system excitability may change when the system underwent transitions in the preictal and postictal processes. Analysis showed that the area of the excitation kernel is positively correlated with the mean firing rate of ictal activity. The kernel duration is also correlated to the amount of ictal activity. The transition into ictal involved the escape from the saddle point foci in the state space trajectory identified using Newton's method.


Asunto(s)
Redes Neurales de la Computación , Convulsiones/fisiopatología , Potenciales de Acción/fisiología , Algoritmos , Animales , Simulación por Computador , Progresión de la Enfermedad , Fenómenos Electrofisiológicos , Epilepsia/fisiopatología , Retroalimentación Fisiológica/fisiología , Hipocampo/fisiología , Técnicas In Vitro , Deficiencia de Magnesio/complicaciones , Deficiencia de Magnesio/fisiopatología , Masculino , Modelos Estadísticos , Técnicas de Placa-Clamp , Ratas , Ratas Wistar , Convulsiones/etiología
13.
Artículo en Inglés | MEDLINE | ID: mdl-38075196

RESUMEN

Glucocorticoids exert pleiotropic effects either by a relatively slow mechanism involving binding to cytosolic/nuclear receptors and regulation of gene expression or by rapid activation of a putative membrane receptor and membrane signal transduction. Rapid glucocorticoid actions are initiated at the membrane and recruit intracellular signaling pathways that engage multiple downstream cellular targets, including lipid and gas intercellular messengers, membrane neurotransmitter receptor trafficking, nuclear glucocorticoid receptor activation and trafficking, and more. Thus, membrane glucocorticoid signaling diverges into a multiplexed array of signaling pathways to simultaneously regulate highly diverse cellular functions, giving these steroid hormones a broad range of rapid regulatory capabilities. In this review, we provide a brief overview of the growing body of knowledge of the cell signaling mechanisms of rapid glucocorticoid actions in the brain.

14.
Nat Commun ; 13(1): 1290, 2022 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-35277502

RESUMEN

Patterned coordination of network activity in the basolateral amygdala (BLA) is important for fear expression. Neuromodulatory systems play an essential role in regulating changes between behavioral states, however the mechanisms underlying this neuromodulatory control of transitions between brain and behavioral states remain largely unknown. We show that chemogenetic Gq activation and α1 adrenoreceptor activation in mouse BLA parvalbumin (PV) interneurons induces a previously undescribed, stereotyped phasic bursting in PV neurons and time-locked synchronized bursts of inhibitory postsynaptic currents and phasic firing in BLA principal neurons. This Gq-coupled receptor activation in PV neurons suppresses gamma oscillations in vivo and in an ex vivo slice model, and facilitates fear memory recall, which is consistent with BLA gamma suppression during conditioned fear expression. Thus, here we identify a neuromodulatory mechanism in PV inhibitory interneurons of the BLA which regulates BLA network oscillations and fear memory recall.


Asunto(s)
Complejo Nuclear Basolateral , Parvalbúminas , Animales , Complejo Nuclear Basolateral/metabolismo , Miedo , Potenciales Postsinápticos Inhibidores/fisiología , Interneuronas/metabolismo , Ratones , Parvalbúminas/metabolismo
15.
Cell Rep ; 41(3): 111509, 2022 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-36261014

RESUMEN

Noradrenergic afferents to hypothalamic corticotropin releasing hormone (CRH) neurons provide a major excitatory drive to the hypothalamic-pituitary-adrenal (HPA) axis via α1 adrenoreceptor activation. Noradrenergic afferents are recruited preferentially by somatic, rather than psychological, stress stimuli. Stress-induced glucocorticoids feed back onto the hypothalamus to negatively regulate the HPA axis, providing a critical autoregulatory constraint that prevents glucocorticoid overexposure and neuropathology. Whether negative feedback mechanisms target stress modality-specific HPA activation is not known. Here, we describe a desensitization of the α1 adrenoreceptor activation of the HPA axis following acute stress in male mice that is mediated by rapid glucocorticoid regulation of adrenoreceptor trafficking in CRH neurons. Glucocorticoid-induced α1 receptor trafficking desensitizes the HPA axis to a somatic but not a psychological stressor. Our findings demonstrate a rapid glucocorticoid suppression of adrenergic signaling in CRH neurons that is specific to somatic stress activation, and they reveal a rapid, stress modality-selective glucocorticoid negative feedback mechanism.


Asunto(s)
Sistema Hipotálamo-Hipofisario , Sistema Hipófiso-Suprarrenal , Animales , Ratones , Masculino , Sistema Hipófiso-Suprarrenal/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Glucocorticoides , Receptores de Glucocorticoides/metabolismo , Estrés Psicológico , Adrenérgicos
16.
J Neurosci ; 30(45): 14980-6, 2010 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-21068301

RESUMEN

Endocannabinoid signaling is distributed throughout the brain, regulating synaptic release of both excitatory and inhibitory neurotransmitters. The presence of endocannabinoid signaling within stress-sensitive nuclei of the hypothalamus, as well as upstream limbic structures such as the amygdala, suggests it may play an important role in regulating the neuroendocrine and behavioral effects of stress. The evidence reviewed here demonstrates that endocannabinoid signaling is involved in both activating and terminating the hypothalamic-pituitary-adrenal axis response to both acute and repeated stress. In addition to neuroendocrine function, however, endocannabinoid signaling is also recruited by stress and glucocorticoid hormones to modulate cognitive and emotional processes such as memory consolidation and extinction. Collectively, these data demonstrate the importance of endocannabinoid signaling at multiple levels as both a regulator and an effector of the stress response.


Asunto(s)
Moduladores de Receptores de Cannabinoides/metabolismo , Endocannabinoides , Hipotálamo/fisiopatología , Estrés Psicológico/fisiopatología , Sinapsis/metabolismo , Transmisión Sináptica/fisiología , Animales , Glucocorticoides/metabolismo , Humanos , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipotálamo-Hipofisario/fisiopatología , Hipotálamo/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Sistema Hipófiso-Suprarrenal/fisiopatología , Estrés Psicológico/metabolismo
17.
Stress ; 14(4): 398-406, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21663538

RESUMEN

Stress activation of the hypothalamic-pituitary-adrenal (HPA) axis culminates in increased circulating corticosteroid concentrations. Stress-induced corticosteroids exert diverse actions in multiple target tissues over a broad range of timescales, ranging from rapid actions, which are induced within seconds to minutes and gene transcription independent, to slow actions, which are delayed, long lasting, and transcription dependent. Rapid corticosteroid actions in the brain include, among others, a fast negative feedback mechanism responsible for shutting down the activated HPA axis centrally. We provide a brief review of the cellular mechanisms responsible for rapid corticosteroid actions in different brain structures of the rat, including the hypothalamus, hippocampus, amygdala, and in the anterior pituitary. We propose a model for the direct feedback inhibition of the HPA axis by glucocorticoids in the hypothalamus. According to this model, glucocorticoids activate membrane glucocorticoid receptors to induce endocannabinoid synthesis in the hypothalamic paraventricular nucleus (PVN) and retrograde cannabinoid type I receptor-mediated suppression of the excitatory synaptic drive to PVN neuroendocrine cells. Rapid corticosteroid actions in the hippocampus, amygdala, and pituitary are mediated by diverse cellular mechanisms and may also contribute to the rapid negative feedback regulation of the HPA neuroendocrine axis as well as to the stress regulation of emotional and spatial memory formation.


Asunto(s)
Glucocorticoides/fisiología , Sistema Hipotálamo-Hipofisario/fisiología , Sistema Hipófiso-Suprarrenal/fisiología , Amígdala del Cerebelo/fisiología , Animales , Moduladores de Receptores de Cannabinoides/biosíntesis , Retroalimentación Fisiológica , Hipocampo/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Neuronas/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Ratas , Receptor Cannabinoide CB1/fisiología , Receptores de Glucocorticoides/fisiología
18.
J Neuroendocrinol ; 33(12): e13061, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34786775

RESUMEN

Oxytocin (OXT) neurons of the hypothalamus are at the center of several physiological functions, including milk ejection, uterus contraction, and maternal and social behavior. In lactating females, OXT neurons show a pattern of burst firing and inter-neuron synchronization during suckling that leads to pulsatile release of surges of OXT into the bloodstream to stimulate milk ejection. This pattern of firing and population synchronization may be facilitated in part by hypothalamic glutamatergic circuits, as has been observed in vitro using brain slices obtained from male rats and neonates. However, it remains unknown how hypothalamic glutamatergic circuits influence OXT cell activity outside the context of lactation. In this review, we summarize the in vivo and in vitro studies that describe the synchronized burst firing pattern of OXT neurons and the implication of hypothalamic glutamate in this pattern of firing. We also make note of the few studies that have traced glutamatergic afferents to the hypothalamic paraventricular and supraoptic nuclei. Finally, we discuss the genetic findings implicating several glutamatergic genes in neurodevelopmental disorders, including autism spectrum disorder, thus underscoring the need for future studies to investigate the impact of these mutations on hypothalamic glutamatergic circuits and the OXT system.


Asunto(s)
Ácido Glutámico/metabolismo , Hipotálamo/metabolismo , Trastornos del Neurodesarrollo/etiología , Neuronas/fisiología , Oxitocina/metabolismo , Animales , Comunicación Celular/fisiología , Femenino , Humanos , Masculino , Red Nerviosa/metabolismo , Red Nerviosa/fisiología , Trastornos del Neurodesarrollo/metabolismo , Trastornos del Neurodesarrollo/fisiopatología , Neuronas/metabolismo , Ratas
19.
J Neurosci ; 29(2): 393-401, 2009 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-19144839

RESUMEN

Glucocorticoids exert an opposing rapid regulation of glutamate and GABA synaptic inputs to hypothalamic magnocellular neurons via the activation of postsynaptic membrane-associated receptors and the release of retrograde messengers. Glucocorticoids suppress synaptic glutamate release via the retrograde release of endocannabinoids and facilitate synaptic GABA release via an unknown retrograde messenger. Here, we show that the glucocorticoid facilitation of GABA inputs is due to the retrograde release of neuronal nitric oxide and that glucocorticoid-induced endocannabinoid synthesis and nitric oxide synthesis are mediated by divergent G-protein signaling mechanisms. While the glucocorticoid-induced, endocannabinoid-mediated suppression of glutamate release is dependent on activation of the G(alpha)s G-protein subunit and cAMP-cAMP-dependent protein kinase activation, the nitric oxide facilitation of GABA release is mediated by G(beta)gamma signaling that leads to activation of neuronal nitric oxide synthase. Our findings indicate, therefore, that glucocorticoids exert opposing rapid actions on glutamate and GABA release by activating divergent G-protein signaling pathways that trigger the synthesis of, and glutamate and GABA synapse-specific retrograde actions of, endocannabinoids and nitric oxide, respectively. The simultaneous rapid stimulation of nitric oxide and endocannabinoid synthesis by glucocorticoids has important implications for the impact of stress on the brain as well as on neural-immune interactions in the hypothalamus.


Asunto(s)
Moduladores de Receptores de Cannabinoides/metabolismo , Inhibidores Enzimáticos/farmacología , Glucocorticoides/farmacología , Ácido Glutámico/metabolismo , Óxido Nítrico/metabolismo , Transducción de Señal/efectos de los fármacos , Sinapsis/efectos de los fármacos , Ácido gamma-Aminobutírico/metabolismo , Animales , Arginina/farmacología , Moduladores de Receptores de Cannabinoides/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Dexametasona/farmacología , Dronabinol/análogos & derivados , Dronabinol/farmacología , Estimulación Eléctrica/métodos , Antagonistas de Aminoácidos Excitadores/farmacología , Proteínas de Unión al GTP/antagonistas & inhibidores , Proteínas de Unión al GTP/metabolismo , Guanosina Difosfato/análogos & derivados , Guanosina Difosfato/farmacología , Hipotálamo/citología , Técnicas In Vitro , Masculino , Neuronas/efectos de los fármacos , Neuronas/fisiología , Donantes de Óxido Nítrico/farmacología , Técnicas de Placa-Clamp/métodos , Penicilamina/análogos & derivados , Penicilamina/farmacología , Piperidinas/farmacología , Piranos/farmacología , Pirazoles/farmacología , Quinoxalinas/farmacología , Ratas , Ratas Sprague-Dawley , Rimonabant , Sinapsis/fisiología , Transmisión Sináptica/fisiología , Tionucleótidos/farmacología , Valina/análogos & derivados , Valina/farmacología
20.
J Physiol ; 588(Pt 6): 939-51, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20123785

RESUMEN

Spike-independent miniature postsynaptic currents are generally stochastic and are therefore not thought to mediate information relay in neuronal circuits. However, we recorded endogenous bursts of IPSCs in hypothalamic magnocellular neurones in the presence of TTX, which implicated a coordinated mechanism of spike-independent GABA release. IPSC bursts were identical in the absence of TTX, although the burst incidence increased 5-fold, indicating that IPSC bursts were composed of miniature IPSCs (mIPSCs), and that the probability of burst generation increased with action potential activity. IPSC bursts required extracellular calcium, although they were not dependent on calcium influx through voltage-gated calcium channels or on calcium mobilization from intracellular stores. Current injections simulating IPSC bursts were capable of triggering and terminating action potential trains. In 25% of dual recordings, a subset of IPSC bursts were highly synchronized in onset in pairs of magnocellular neurones. Synchronized IPSC bursts displayed properties that were consistent with simultaneous release at GABA synapses shared between pairs of postsynaptic magnocellular neurones. Synchronized bursts of inhibitory synaptic inputs represent a novel mechanism that may contribute to the action potential burst generation, termination and synchronization responsible for pulsatile hormone release from neuroendocrine cells.


Asunto(s)
Potenciales de Acción/fisiología , Potenciales Postsinápticos Inhibidores/fisiología , Neuronas/fisiología , Ácido gamma-Aminobutírico/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Calcio/metabolismo , Canales de Calcio/fisiología , Fenómenos Electrofisiológicos/fisiología , Hipotálamo/citología , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Masculino , Neuronas/citología , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , Ratas , Ratas Wistar , Bloqueadores de los Canales de Sodio/farmacología , Tetrodotoxina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA