Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Curr Biol ; 33(24): 5381-5389.e4, 2023 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-37992720

RESUMEN

Endotherms can survive low temperatures and food shortage by actively entering a hypometabolic state known as torpor. Although the decrease in metabolic rate and body temperature (Tb) during torpor is controlled by the brain, the specific neural circuits underlying these processes have not been comprehensively elucidated. In this study, we identify the neural circuits involved in torpor regulation by combining whole-brain mapping of torpor-activated neurons, cell-type-specific manipulation of neural activity, and viral tracing-based circuit mapping. We find that Trpm2-positive neurons in the preoptic area and Vgat-positive neurons in the dorsal medial hypothalamus are activated during torpor. Genetic silencing shows that the activity of either cell type is necessary to enter the torpor state. Finally, we show that these cells receive projections from the arcuate and suprachiasmatic nucleus and send projections to brain regions involved in thermoregulation. Our results demonstrate an essential role of hypothalamic neurons in the regulation of Tb and metabolic rate during torpor and identify critical nodes of the torpor regulatory network.


Asunto(s)
Hipotálamo , Letargo , Hipotálamo/fisiología , Letargo/fisiología , Área Preóptica , Núcleo Supraquiasmático , Encéfalo
2.
J Physiol ; 600(4): 815-827, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-33899241

RESUMEN

KEY POINTS: Melanin-concentrating hormone (MCH) neuron-ablated mice exhibit increased energy expenditure and reduced fat weight. Increased brown adipose tissue (BAT) activity and locomotor activity-independent energy expenditure contributed to body weight reduction in MCH neuron-ablated mice. MCH neurons send inhibitory input to the medullary raphe nucleus to modulate BAT activity. ABSTRACT: Hypothalamic melanin-concentrating hormone (MCH) peptide robustly affects energy homeostasis. However, it is unclear whether and how MCH-producing neurons, which contain and release a variety of neuropeptides/transmitters, regulate energy expenditure in the central nervous system and peripheral tissues. We thus examined the regulation of energy expenditure by MCH neurons, focusing on interscapular brown adipose tissue (BAT) activity. MCH neuron-ablated mice exhibited reduced body weight, increased oxygen consumption, and increased BAT activity, which improved locomotor activity-independent energy expenditure. Trans-neuronal retrograde tracing with the recombinant pseudorabies virus revealed that MCH neurons innervate BAT via the sympathetic premotor region in the medullary raphe nucleus (MRN). MRN neurons were activated by MCH neuron ablation. Therefore, endogenous MCH neuron activity negatively modulates energy expenditure via BAT inhibition. MRN neurons might receive inhibitory input from MCH neurons to suppress BAT activity.


Asunto(s)
Tejido Adiposo Pardo , Hormonas Hipotalámicas , Tejido Adiposo Pardo/metabolismo , Animales , Metabolismo Energético , Hormonas Hipotalámicas/metabolismo , Hipotálamo/fisiología , Melaninas/metabolismo , Ratones , Neuronas/fisiología , Hormonas Hipofisarias/metabolismo
3.
Mol Brain ; 14(1): 104, 2021 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-34193206

RESUMEN

The perifornical area of the hypothalamus has been known as the center for the defense response, or fight-or-flight response, which is characterized by a concomitant rise in arterial blood pressure, heart rate, and respiratory frequency. It is well established that orexin neurons, which are located in this region, play a critical role in this response. In this study, we further examined this role by recording orexin neuronal activity and heart rate in freely moving mice using an original dual-channel fiber photometry system in vivo. Analysis of orexin neuron activity in relation to autonomic responses to aversive stimuli revealed a rapid increase in neuronal activity just prior to changes in heart rate. In addition, we examined whether orexin neurons would be activated by a conditioned neutral sound that was previously associated with aversive stimulus. We show that the memory of the aversive stimulus activated orexin neurons and increased heart rate. Our data suggest that orexin neurons are a key component linking aversive emotions to autonomic defense response. Our data also suggest that targeting orexin neurons may enable treatment of psychiatric disorders associated with chronic stress and traumatic memories.


Asunto(s)
Emociones/fisiología , Frecuencia Cardíaca/fisiología , Movimiento/fisiología , Neuronas/metabolismo , Orexinas/metabolismo , Estimulación Acústica , Animales , Temperatura Corporal , Ratones Transgénicos , Fotometría , Tiempo de Reacción , Estrés Fisiológico
4.
Sci Rep ; 10(1): 13639, 2020 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-32788592

RESUMEN

The stress response is a physiological system for adapting to various internal and external stimuli. Corticotropin-releasing factor-producing neurons in the paraventricular nucleus of the hypothalamus (PVN-CRF neurons) are known to play an important role in the stress response as initiators of the hypothalamic-pituitary-adrenal axis. However, the mechanism by which activity of PVN-CRF neurons is regulated by other neurons and bioactive substances remains unclear. Here, we developed a screening method using calcium imaging to identify how physiological substances directly affect the activity of PVN-CRF neurons. We used acute brain slices expressing a genetically encoded calcium indicator in PVN-CRF neurons using CRF-Cre recombinase mice and an adeno-associated viral vector under Cre control. PVN-CRF neurons were divided into ventral and dorsal portions. Bath application of candidate substances revealed 12 substances that increased and 3 that decreased intracellular calcium concentrations. Among these substances, angiotensin II and histamine mainly increased calcium in the ventral portion of the PVN-CRF neurons via AT1 and H1 receptors, respectively. Conversely, carbachol mainly increased calcium in the dorsal portion of the PVN-CRF neurons via both nicotinic and muscarinic acetylcholine receptors. Our method provides a precise and reliable means of evaluating the effect of a substance on PVN-CRF neuronal activity.


Asunto(s)
Calcio/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Hipotálamo/metabolismo , Neuronas/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Angiotensina II/farmacología , Animales , Hipotálamo/efectos de los fármacos , Ratones , Neuronas/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Vasoconstrictores/farmacología
5.
Cell Rep ; 24(8): 2191-2195.e4, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-30134178

RESUMEN

To understand brain circuits of cognitive behaviors under natural conditions, we developed techniques for imaging neuronal activities from large neuronal populations in the deep layer cortex of the naturally behaving common marmoset. Animals retrieved food pellets or climbed ladders as a miniature fluorescence microscope monitored hundreds of calcium indicator-expressing cortical neurons in the right primary motor cortex. This technique, which can be adapted to other brain regions, can deepen our understanding of brain circuits by facilitating longitudinal population analyses of neuronal representation associated with cognitive naturalistic behaviors and their pathophysiological processes.


Asunto(s)
Conducta Animal/fisiología , Calcio/metabolismo , Corteza Motora/fisiología , Neuronas/fisiología , Animales , Haplorrinos
6.
Neurosci Res ; 118: 74-81, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28526553

RESUMEN

Sleep is one of the most important physiological functions in mammals. It is regulated by not only homeostatic regulation but also circadian clock. Several neuropeptide-producing neurons located in the hypothalamus are implicated in the regulation of sleep/wakefulness. Among them, orexin/hypocretin-producing neurons (orexin neurons) are a crucial component for maintenance of wakefulness, because lack of orexin function results in narcolepsy, which is a sleep disorder. Recent findings have identified substances that excite or inhibit neural activity of orexin neurons. Furthermore neural projections of the neurons which release these substances have been revealed. In addition to orexin, melanin concentrating hormone (MCH)-producing neurons in the lateral hypothalamic area (LHA) are also implicated in the regulation of sleep/wakefulness. MCH neurons are active during sleep but become silent during wakefulness. Recently developed innovative methods including optogenetics and pharmacogenetics have provided substantial insights into the regulation of sleep/wakefulness. In vivo optical recordings and retrograde and anterograde tracing methods will allow us to understand additional details regarding important interactions between these two types of neurons in the LHA and other neurons in the brain. Finally we discuss the circadian clock and sleep/wake cycle. Understanding of the neural networks and its circadian modulation of sleep/wake cycles remain to be investigated.


Asunto(s)
Hormonas Hipotalámicas/fisiología , Hipotálamo/fisiología , Melaninas/fisiología , Orexinas/fisiología , Hormonas Hipofisarias/fisiología , Sueño/fisiología , Vigilia/fisiología , Animales , Humanos
7.
Am J Physiol Heart Circ Physiol ; 312(4): H808-H817, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-28159808

RESUMEN

Orexin neurons, and activation of orexin receptors, are generally thought to be sympathoexcitatory; however, the functional connectivity between orexin neurons and a likely sympathetic target, the hypothalamic spinally projecting neurons (SPNs) in the paraventricular nucleus of the hypothalamus (PVN) has not been established. To test the hypothesis that orexin neurons project directly to SPNs in the PVN, channelrhodopsin-2 (ChR2) was selectively expressed in orexin neurons to enable photoactivation of ChR2-expressing fibers while examining evoked postsynaptic currents in SPNs in rat hypothalamic slices. Selective photoactivation of orexin fibers elicited short-latency postsynaptic currents in all SPNs tested (n = 34). These light-triggered responses were heterogeneous, with a majority being excitatory glutamatergic responses (59%) and a minority of inhibitory GABAergic (35%) and mixed glutamatergic and GABAergic currents (6%). Both glutamatergic and GABAergic responses were present in the presence of tetrodotoxin and 4-aminopyridine, suggesting a monosynaptic connection between orexin neurons and SPNs. In addition to generating postsynaptic responses, photostimulation facilitated action potential firing in SPNs (current clamp configuration). Glutamatergic, but not GABAergic, postsynaptic currents were diminished by application of the orexin receptor antagonist almorexant, indicating orexin release facilitates glutamatergic neurotransmission in this pathway. This work identifies a neuronal circuit by which orexin neurons likely exert sympathoexcitatory control of cardiovascular function.NEW & NOTEWORTHY This is the first study to establish, using innovative optogenetic approaches in a transgenic rat model, that there are robust heterogeneous projections from orexin neurons to paraventricular spinally projecting neurons, including excitatory glutamatergic and inhibitory GABAergic neurotransmission. Endogenous orexin release modulates glutamatergic, but not GABAergic, neurotransmission in these pathways.


Asunto(s)
Hipotálamo/citología , Hipotálamo/metabolismo , Neuronas/metabolismo , Orexinas/metabolismo , Núcleo Hipotalámico Paraventricular/citología , Núcleo Hipotalámico Paraventricular/metabolismo , Médula Espinal/citología , Médula Espinal/metabolismo , Acetamidas/farmacología , Potenciales de Acción/fisiología , Animales , Animales Modificados Genéticamente , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Ácido Glutámico/metabolismo , Hipotálamo/diagnóstico por imagen , Isoquinolinas/farmacología , Masculino , Vías Nerviosas/citología , Vías Nerviosas/diagnóstico por imagen , Optogenética , Receptores de Orexina/genética , Receptores de Orexina/metabolismo , Orexinas/genética , Núcleo Hipotalámico Paraventricular/diagnóstico por imagen , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Médula Espinal/diagnóstico por imagen , Ácido gamma-Aminobutírico/metabolismo
8.
Sci Rep ; 6: 36039, 2016 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-27824065

RESUMEN

Orexin/hypocretin neurons play a crucial role in the regulation of sleep/wakefulness, primarily in the maintenance of wakefulness. These neurons innervate wide areas of the brain and receive diverse synaptic inputs including those from serotonergic (5-HT) neurons in the raphe nucleus. Previously we showed that pharmacological application of 5-HT directly inhibited orexin neurons via 5-HT1A receptors. However, it was still unclear how 5-HT neurons regulated orexin neurons since 5-HT neurons contain not only 5-HT but also other neurotransmitters. To reveal this, we generated new triple transgenic mice in which orexin neurons express enhanced green fluorescent protein (EGFP) and 5-HT neurons express channelrhodopsin2 (ChR2). Immunohistochemical studies show that nerve endings of ChR2-expressing 5-HT neurons are in close apposition to EGFP-expressing orexin neurons in the lateral hypothalamic area. Using these mice, we could optogenetically activate 5-HT nerve terminals and record postsynaptic effects from orexin neurons. Activation of nerve terminals of 5-HT neurons directly inhibited orexin neurons via the 5HT1A receptor, and also indirectly inhibited orexin neurons by facilitating GABAergic inhibitory inputs without affecting glutamatergic inputs. Increased GABAergic inhibitory inputs in orexin neurons were confirmed by the pharmacological application of 5-HT. These results suggest that orexin neurons are inhibited by 5-HT neurons, primarily via 5-HT, in both direct and indirect manners.


Asunto(s)
Neuronas GABAérgicas/fisiología , Hipotálamo/fisiología , Inhibición Neural , Neuronas Serotoninérgicas/fisiología , Animales , Ratones Transgénicos , Optogenética , Vigilia
9.
Neuroscience ; 339: 47-53, 2016 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-27693474

RESUMEN

Orexin neurons are known to augment the sympathetic control of cardiovascular function, however the role of orexin neurons in parasympathetic cardiac regulation remains unclear. To test the hypothesis that orexin neurons contribute to parasympathetic control we selectively expressed channelrhodopsin-2 (ChR2) in orexin neurons in orexin-Cre transgenic rats and examined postsynaptic currents in cardiac vagal neurons (CVNs) in the dorsal motor nucleus of the vagus (DMV). Simultaneous photostimulation and recording in ChR2-expressing orexin neurons in the lateral hypothalamus resulted in reliable action potential firing as well as large whole-cell currents suggesting a strong expression of ChR2 and reliable optogenetic excitation. Photostimulation of ChR2-expressing fibers in the DMV elicited short-latency (ranging from 3.2ms to 8.5ms) postsynaptic currents in 16 out of 44 CVNs tested. These responses were heterogeneous and included excitatory glutamatergic (63%) and inhibitory GABAergic (37%) postsynaptic currents. The results from this study suggest different sub-population of orexin neurons may exert diverse influences on brainstem CVNs and therefore may play distinct functional roles in parasympathetic control of the heart.


Asunto(s)
Tronco Encefálico/metabolismo , Hipotálamo/metabolismo , Neuronas/metabolismo , Orexinas/metabolismo , Nervio Vago/metabolismo , Potenciales de Acción/fisiología , Animales , Tronco Encefálico/citología , Femenino , Ácido Glutámico/metabolismo , Hipotálamo/citología , Masculino , Vías Nerviosas/citología , Vías Nerviosas/metabolismo , Neuronas/citología , Optogenética , Ratas Transgénicas , Potenciales Sinápticos/fisiología , Técnicas de Cultivo de Tejidos , Nervio Vago/citología , Ácido gamma-Aminobutírico/metabolismo
10.
J Neurophysiol ; 116(5): 2250-2259, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27559138

RESUMEN

Evidence of impaired function of orexin neurons has been found in individuals with cardiorespiratory disorders, such as obstructive sleep apnea (OSA) and sudden infant death syndrome (SIDS), but the mechanisms responsible are unknown. Individuals with OSA and SIDS experience repetitive breathing cessations and/or rebreathing of expired air, resulting in hypoxia/hypercapnia (H/H). In this study, we examined the responses of fluorescently identified rat orexin neurons in the lateral hypothalamus to acute H/H to test if and how these neurons alter their activity and function during this challenge. Experiments were conducted in an in vitro slice preparation using voltage-clamp and current-clamp configurations. H/H (10 min) induced hyperpolarization, accompanied by rapid depression, and finally, cessation of firing activity in orexin neurons. Hypoxia alone had similar but less potent effects. H/H did not alter the frequency of inhibitory glycinergic postsynaptic currents. The frequency of GABAergic currents was diminished but only at 8-10 min of H/H. In contrast, the frequency of excitatory glutamatergic postsynaptic events was diminished as early as 2-4 min of H/H. In the presence of glutamatergic receptor blockers, the inhibitory effects of H/H on the firing activity and membrane potential of orexin neurons persisted but to a lesser extent. In conclusion, both direct alteration of postsynaptic membrane properties and diminished glutamatergic neurotransmission likely contribute to the inhibition of orexin neurons by H/H. These mechanisms could be responsible for the decreased function of orexin in individuals at risk for OSA and SIDS.


Asunto(s)
Hipotálamo/metabolismo , Neuronas/metabolismo , Receptores de Orexina/biosíntesis , Consumo de Oxígeno/fisiología , Animales , Hipoxia de la Célula/fisiología , Hipercapnia/metabolismo , Hipotálamo/química , Potenciales de la Membrana/fisiología , Neuronas/química , Receptores de Orexina/análisis , Orexinas/análisis , Orexinas/antagonistas & inhibidores , Orexinas/biosíntesis , Técnicas de Cultivo de Órganos , Ratas , Ratas Transgénicas
11.
Nagoya J Med Sci ; 75(1-2): 29-36, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23544265

RESUMEN

Orexins, also known as hypocretins, are neuropeptides that are exclusively expressed by neurons in the lateral hypothalamic area. Although originally recognized as regulators of feeding behavior, orexins are now mainly regarded as key modulators of the sleep/wakefulness cycle. In addition, anatomical studies of neural networks and analyses of transgenic mice have revealed integrated roles for orexin neurons in the coordination of emotion, energy homeostasis, and the reward system. A functional link between the limbic system and orexin neurons may be important for increasing vigilance in response to emotional stimuli. These findings suggest that orexin neurons relay information about an organism's environment to maintain the proper amount of sleep and wakefulness in animals.


Asunto(s)
Hipotálamo/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neuropéptidos/metabolismo , Sueño , Vigilia , Animales , Humanos , Neuronas/metabolismo , Receptores de Orexina , Orexinas , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropéptido/metabolismo , Transducción de Señal
12.
J Physiol ; 574(Pt 2): 399-414, 2006 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16627567

RESUMEN

Hypocretin/orexin (Hcrt) is a critical neurotransmitter for the maintenance of wakefulness and has been implicated in several other functions, including energy metabolism and reward. Using whole-cell patch-clamp recordings from transgenic mice in which enhanced green fluorescent protein was linked to the Hcrt promoter, we investigated GABAergic control of the Hcrt neurones in hypothalamic slices. Bath application of GABA or muscimol caused an early hyperpolarization mediated by Cl(-) and a late depolarization mediated by the efflux of bicarbonate. These GABA(A) receptor-mediated responses were blocked by picrotoxin and bicuculline. Under the GABA(A) blockade condition, GABA produced consistent hyperpolarization, decreased firing rate and input resistance. The selective GABA(B) agonist (R)-baclofen caused a similar response with an EC(50) of 7.1 mum. The effects of (R)-baclofen were blocked by the GABA(B) antagonist CGP 52432 but persisted in the presence of tetrodotoxin, suggesting direct postsynaptic effects. The existence of GABA(B) modulation was supported by GABA(B(1)) subunit immunoreactivity on Hcrt cells colabelled with antisera to the Hcrt-2 peptide. Furthermore, GABA(B) receptor activation inhibited the presynaptic release of both glutamate and GABA. (R)-Baclofen depressed the amplitude of evoked excitatory postsynaptic currents (EPSCs) and inhibitory synaptic currents (IPSCs), and also decreased the frequency of both spontaneous and miniature EPSCs and IPSCs with a modest effect on their amplitudes. These data suggest that GABA(B) receptors modulate Hcrt neuronal activity via both pre- and postsynaptic mechanisms, which may underlie the promotion of non-rapid eye movement sleep and have implications for the use of GABA(B) agonists in the treatment of substance addiction through direct interaction with the Hcrt system.


Asunto(s)
Hipotálamo/fisiología , Péptidos y Proteínas de Señalización Intracelular/análisis , Neuronas/química , Neuronas/fisiología , Neuropéptidos/análisis , Receptores de GABA-B/fisiología , Potenciales de Acción/fisiología , Animales , Agonistas del GABA/farmacología , Antagonistas del GABA/farmacología , Antagonistas de Receptores de GABA-A , Antagonistas de Receptores de GABA-B , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Orexina , Orexinas , Receptores Acoplados a Proteínas G , Receptores de GABA-A/efectos de los fármacos , Receptores de GABA-A/fisiología , Receptores de GABA-B/efectos de los fármacos , Receptores de Neuropéptido , Sueño/fisiología , Sinapsis/efectos de los fármacos , Sinapsis/fisiología , Vigilia/fisiología
13.
Endocrinology ; 147(5): 2306-14, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16484324

RESUMEN

Ghrelin, an acylated peptide produced predominantly in the stomach, stimulates feeding and GH secretion via interactions with the GH secretagogue type 1a receptor (GHS-R1a), the functionally active form of the GHS-R. Ghrelin molecules exist in the stomach and hypothalamus as two major endogenous forms, a form acylated at serine 3 (ghrelin) and a des-acylated form (des-acyl ghrelin). Acylation is indispensable for the binding of ghrelin to the GHS-R1a. Ghrelin enhances feeding via the neuronal pathways of neuropeptide Y and orexin, which act as orexigenic peptides in the hypothalamus. We here studied the effect of des-acyl ghrelin on feeding behavior. Intracerebroventricular (icv) administration of rat des-acyl ghrelin to rats or mice fed ad libitum stimulated feeding during the light phase; neither ip nor icv administration of des-acyl ghrelin to fasting mice suppressed feeding. The icv administration of des-acyl ghrelin induced the expression of Fos, a marker of neuronal activation, in orexin-expressing neurons of the lateral hypothalamic area, but not neuropeptide Y-expressing neurons of the arcuate nucleus. Peripheral administration of des-acyl ghrelin to rats or mice did not affect feeding. Although icv administration of ghrelin did not induce food intake in GHS-R-deficient mice, it did in orexin-deficient mice. In contrast, icv administration of des-acyl ghrelin stimulated feeding in GHS-R-deficient mice, but not orexin-deficient mice. Des-acyl ghrelin increased the intracellular calcium concentrations in isolated orexin neurons. Central des-acyl ghrelin may activate orexin-expressing neurons, perhaps functioning in feeding regulation through interactions with a target protein distinct from the GHS-R.


Asunto(s)
Conducta Alimentaria , Hormonas Peptídicas/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Animales , Calcio/metabolismo , Cromatografía Líquida de Alta Presión , Citosol/metabolismo , Mucosa Gástrica/metabolismo , Ghrelina , Hormona del Crecimiento/metabolismo , Hipotálamo/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Movimiento , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Neuropéptidos/metabolismo , Receptores de Orexina , Orexinas , Hormonas Peptídicas/metabolismo , Péptidos/química , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Receptores de Ghrelina , Receptores de Neuropéptido , Factores de Tiempo
14.
Neuron ; 46(2): 297-308, 2005 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-15848807

RESUMEN

The finding of orexin/hypocretin deficiency in narcolepsy patients suggests that this hypothalamic neuropeptide plays a crucial role in regulating sleep/wakefulness states. However, very little is known about the synaptic input of orexin/hypocretin-producing neurons (orexin neurons). We applied a transgenic method to map upstream neuronal populations that have synaptic connections to orexin neurons and revealed that orexin neurons receive input from several brain areas. These include the amygdala, basal forebrain cholinergic neurons, GABAergic neurons in the preoptic area, and serotonergic neurons in the median/paramedian raphe nuclei. Monoamine-containing groups that are innervated by orexin neurons do not receive reciprocal connections, while cholinergic neurons in the basal forebrain have reciprocal connections, which might be important for consolidating wakefulness. Electrophysiological study showed that carbachol excites almost one-third of orexin neurons and inhibits a small population of orexin neurons. These neuroanatomical findings provide important insights into the neural pathways that regulate sleep/wakefulness states.


Asunto(s)
Hipotálamo/anatomía & histología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Vías Nerviosas/anatomía & histología , Neuronas/citología , Neuropéptidos/metabolismo , Animales , Tronco Encefálico/anatomía & histología , Tronco Encefálico/efectos de los fármacos , Tronco Encefálico/ultraestructura , Carbacol/farmacología , Agonistas Colinérgicos/farmacología , Proteínas Fluorescentes Verdes/genética , Humanos , Hipotálamo/efectos de los fármacos , Hipotálamo/ultraestructura , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Transgénicos , Vías Nerviosas/efectos de los fármacos , Neuronas/fisiología , Orexinas , Técnicas de Placa-Clamp , Proteínas Recombinantes de Fusión/genética , Tetrodotoxina/genética , Vigilia/fisiología
15.
J Neurosci ; 24(50): 11439-48, 2004 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-15601950

RESUMEN

The hypothalamic neuropeptides hypocretins (orexins) play a crucial role in the stability of arousal and alertness. We tested whether the hypocretinergic system is a critical component of the stress response activated by the corticotropin-releasing factor (CRF). Our results show that CRF-immunoreactive terminals make direct contact with hypocretin-expressing neurons in the lateral hypothalamus and that numerous hypocretinergic neurons express the CRF-R1/2 receptors. We also demonstrate that application of CRF to hypothalamic slices containing identified hypocretin neurons depolarizes membrane potential and increases firing rate in a subpopulation of hypocretinergic cells. CRF-induced depolarization was tetrodotoxin insensitive and was blocked by the peptidergic CRF-R1 antagonist astressin. Moreover, activation of hypocretinergic neurons in response to acute stress was severely impaired in CRF-R1 knock-out mice. Together, our data provide evidence of a direct neuroanatomical and physiological input from CRF peptidergic system onto hypocretin neurons. We propose that, after stressor stimuli, CRF stimulates the release of hypocretins and that this circuit contributes to activation and maintenance of arousal associated with the stress response.


Asunto(s)
Nivel de Alerta/fisiología , Química Encefálica/fisiología , Hormona Liberadora de Corticotropina/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Neuropéptidos/fisiología , Estrés Fisiológico/fisiopatología , Potenciales de Acción/fisiología , Animales , Encéfalo/anatomía & histología , Hormona Liberadora de Corticotropina/análisis , Femenino , Hipotálamo/química , Hipotálamo/fisiología , Inmunohistoquímica , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intracelular/análisis , Masculino , Potenciales de la Membrana/fisiología , Ratones , Ratones Noqueados , Vías Nerviosas/anatomía & histología , Vías Nerviosas/química , Neuronas/química , Neuronas/fisiología , Neuropéptidos/análisis , Receptores de Orexina , Orexinas , Receptores de Hormona Liberadora de Corticotropina/genética , Receptores de Hormona Liberadora de Corticotropina/fisiología , Receptores Acoplados a Proteínas G , Receptores de Neuropéptido , Proteínas Recombinantes de Fusión
16.
J Neurosci ; 24(32): 7159-66, 2004 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-15306649

RESUMEN

Both orexin and serotonin (5-HT) have important roles in the regulation of sleep-wakefulness, as well as in feeding behavior. We examined the effects of 5-HT on orexin/hypocretin neurons, using hypothalamic slices prepared from orexin/enhanced green fluorescent protein (EGFP) transgenic mice in which EGFP is expressed exclusively in orexin neurons. Patch-clamp recording from EGFP-expressing cells showed that 5-HT hyperpolarized all orexin neurons in a concentration-dependent manner. The response was inhibited by the 5-HT1A receptor antagonist WAY100635. A 5-HT1A receptor agonist, 8-hydroxy-2-(dl-N-propyl-amino)tetralin, also evoked hyperpolarization on orexin neurons with potency comparable with 5-HT. A low concentration of Ba2+ (30 microM) inhibited 5-HT-induced hyperpolarization. Single-channel recording revealed that the conductance of 5-HT-induced channel activity was 33.8 pS, which is in good agreement with that of the G-protein-coupled inward rectifier potassium channel (GIRK). Moreover, 5-HT1A receptor-like immunoreactivity was observed on orexin neurons, and 5-HT transporter immunoreactive nerve endings are in close apposition to orexin neurons. Intracerebroventricular injection of the 5-HT1A receptor-selective antagonist WAY100635 (100 ng) increased locomotor activity during the latter half of dark phase in wild-type mice but not in orexin/ataxin-3 mice in which orexin neurons are specifically ablated, suggesting that activation of orexin neurons is necessary for the WAY100635-induced increase in locomotor activity. These results indicate that 5-HT hyperpolarizes orexin neurons through the 5-HT1A receptor and subsequent activation of the GIRK and that this inhibitory serotonergic input to the orexin neurons is likely to be important for the physiological regulation of this neuropeptide system.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neuronas/fisiología , Neuropéptidos/metabolismo , Receptor de Serotonina 5-HT1A/fisiología , Animales , Ataxina-3 , Femenino , Canales de Potasio Rectificados Internamente Asociados a la Proteína G , Proteínas Fluorescentes Verdes/genética , Hipotálamo/citología , Hipotálamo/metabolismo , Inmunohistoquímica , Técnicas In Vitro , Inyecciones Intraventriculares , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Glicoproteínas de Membrana/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Neuropéptidos/genética , Proteínas Nucleares , Receptores de Orexina , Orexinas , Técnicas de Placa-Clamp , Canales de Potasio de Rectificación Interna/agonistas , Receptor de Serotonina 5-HT1A/biosíntesis , Receptores Acoplados a Proteínas G , Receptores de Neuropéptido , Proteínas Represoras , Serotonina/farmacología , Antagonistas de la Serotonina/administración & dosificación , Antagonistas de la Serotonina/farmacología , Proteínas de Transporte de Serotonina en la Membrana Plasmática , Agonistas de Receptores de Serotonina/farmacología , Tetrodotoxina/farmacología , Factores de Transcripción
17.
Eur J Neurosci ; 19(6): 1524-34, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15066149

RESUMEN

Orexin-A and -B (hypocretin-1 and -2) have been implicated in the stimulation of feeding. Here we show the effector neurons and signaling mechanisms for the orexigenic action of orexins in rats. Immunohistochemical methods showed that orexin axon terminals contact with neuropeptide Y (NPY)- and proopiomelanocortin (POMC)-positive neurons in the arcuate nucleus (ARC) of the rats. Microinjection of orexins into the ARC markedly increased food intake. Orexins increased cytosolic Ca(2+) concentration ([Ca(2+)](i)) in the isolated neurons from the ARC, which were subsequently shown to be immunoreactive for NPY. The increases in [Ca(2+)](i) were inhibited by blockers of phospholipase C (PLC), protein kinase C (PKC) and Ca(2+) uptake into endoplasmic reticulum. The stimulation of food intake and increases in [Ca(2+)](i) in NPY neurons were greater with orexin-A than with orexin-B, indicative of involvement of the orexin-1 receptor (OX(1)R). In contrast, orexin-A and -B equipotently attenuated [Ca(2+)](i) oscillations and decreased [Ca(2+)](i) levels in POMC-containing neurons. These effects were counteracted by pertussis toxin, suggesting involvement of the orexin-2 receptor and Gi/Go subtypes of GTP-binding proteins. Orexins also decreased [Ca(2+)](i) levels in glucose-responsive neurons in the ventromedial hypothalamus (VMH), a satiety center. Leptin exerted opposite effects on these three classes of neurons. These results demonstrate that orexins directly regulate NPY, POMC and glucose-responsive neurons in the ARC and VMH, in a manner reciprocal to leptin. Orexin-A evokes Ca(2+) signaling in NPY neurons via OX(1)R-PLC-PKC and IP(3) pathways. These neural pathways and intracellular signaling mechanisms may play key roles in the orexigenic action of orexins.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Proteínas Portadoras/farmacología , Glucosa/metabolismo , Hipotálamo/citología , Péptidos y Proteínas de Señalización Intracelular , Leptina/farmacología , Neuropéptido Y/metabolismo , Neuropéptidos/farmacología , Proopiomelanocortina/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Ingestión de Alimentos/efectos de los fármacos , Inhibidores Enzimáticos , Fura-2/metabolismo , Inmunohistoquímica , Modelos Neurológicos , Neuronas/clasificación , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Orexinas , Toxina del Pertussis/farmacología , Ratas , Ratas Sprague-Dawley , Ácido gamma-Aminobutírico/farmacología
18.
Neuron ; 38(5): 701-13, 2003 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-12797956

RESUMEN

Mammals respond to reduced food availability by becoming more wakeful and active, yet the central pathways regulating arousal and instinctual motor programs (such as food seeking) according to homeostatic need are not well understood. We demonstrate that hypothalamic orexin neurons monitor indicators of energy balance and mediate adaptive augmentation of arousal in response to fasting. Activity of isolated orexin neurons is inhibited by glucose and leptin and stimulated by ghrelin. Orexin expression of normal and ob/ob mice correlates negatively with changes in blood glucose, leptin, and food intake. Transgenic mice, in which orexin neurons are ablated, fail to respond to fasting with increased wakefulness and activity. These findings indicate that orexin neurons provide a crucial link between energy balance and arousal.


Asunto(s)
Nivel de Alerta/genética , Metabolismo Energético/genética , Privación de Alimentos/fisiología , Hambre/fisiología , Hipotálamo/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Neuronas/metabolismo , Neuropéptidos/deficiencia , Animales , Nivel de Alerta/efectos de los fármacos , Glucemia/efectos de los fármacos , Glucemia/fisiología , Proteínas Portadoras/genética , Metabolismo Energético/efectos de los fármacos , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Espacio Extracelular/metabolismo , Ghrelina , Glucosa/metabolismo , Glucosa/farmacología , Proteínas Fluorescentes Verdes , Homeostasis/efectos de los fármacos , Homeostasis/genética , Hambre/efectos de los fármacos , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Leptina/metabolismo , Leptina/farmacología , Proteínas Luminiscentes , Masculino , Potenciales de la Membrana/genética , Ratones , Ratones Transgénicos , Neuronas/citología , Neuronas/efectos de los fármacos , Neuropéptidos/genética , Orexinas , Técnicas de Cultivo de Órganos , Hormonas Peptídicas/metabolismo , Hormonas Peptídicas/farmacología , Proteínas Recombinantes de Fusión , Transmisión Sináptica/genética , Transgenes/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA