Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
1.
Science ; 377(6601): 63-72, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35771921

RESUMEN

In mice, social defeat stress (SDS), an ethological model for psychosocial stress, induces sleep. Such sleep could enable resilience, but how stress promotes sleep is unclear. Activity-dependent tagging revealed a subset of ventral tegmental area γ-aminobutyric acid (GABA)-somatostatin (VTAVgat-Sst) cells that sense stress and drive non-rapid eye movement (NREM) and REM sleep through the lateral hypothalamus and also inhibit corticotropin-releasing factor (CRF) release in the paraventricular hypothalamus. Transient stress enhances the activity of VTAVgat-Sst cells for several hours, allowing them to exert their sleep effects persistently. Lesioning of VTAVgat-Sst cells abolished SDS-induced sleep; without it, anxiety and corticosterone concentrations remained increased after stress. Thus, a specific circuit allows animals to restore mental and body functions by sleeping, potentially providing a refined route for treating anxiety disorders.


Asunto(s)
Resiliencia Psicológica , Sueño , Derrota Social , Estrés Psicológico , Área Tegmental Ventral , Animales , Hormona Liberadora de Corticotropina/metabolismo , Área Hipotalámica Lateral/fisiopatología , Ratones , Sueño REM , Somatostatina/metabolismo , Estrés Psicológico/fisiopatología , Área Tegmental Ventral/fisiopatología , Ácido gamma-Aminobutírico/metabolismo
2.
Science ; 375(6583): eabh3021, 2022 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-35201886

RESUMEN

Sleep quality declines with age; however, the underlying mechanisms remain elusive. We found that hyperexcitable hypocretin/orexin (Hcrt/OX) neurons drive sleep fragmentation during aging. In aged mice, Hcrt neurons exhibited more frequent neuronal activity epochs driving wake bouts, and optogenetic activation of Hcrt neurons elicited more prolonged wakefulness. Aged Hcrt neurons showed hyperexcitability with lower KCNQ2 expression and impaired M-current, mediated by KCNQ2/3 channels. Single-nucleus RNA-sequencing revealed adaptive changes to Hcrt neuron loss in the aging brain. Disruption of Kcnq2/3 genes in Hcrt neurons of young mice destabilized sleep, mimicking aging-associated sleep fragmentation, whereas the KCNQ-selective activator flupirtine hyperpolarized Hcrt neurons and rejuvenated sleep architecture in aged mice. Our findings demonstrate a mechanism underlying sleep instability during aging and a strategy to improve sleep continuity.


Asunto(s)
Envejecimiento , Neuronas/fisiología , Orexinas/fisiología , Privación de Sueño/fisiopatología , Sueño , Vigilia , Aminopiridinas/farmacología , Animales , Sistemas CRISPR-Cas , Electroencefalografía , Electromiografía , Femenino , Área Hipotalámica Lateral/fisiopatología , Canal de Potasio KCNQ2/genética , Canal de Potasio KCNQ2/metabolismo , Canal de Potasio KCNQ3/genética , Canal de Potasio KCNQ3/metabolismo , Masculino , Ratones , Narcolepsia/genética , Narcolepsia/fisiopatología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Vías Nerviosas , Optogenética , Técnicas de Placa-Clamp , RNA-Seq , Calidad del Sueño
3.
FASEB J ; 35(9): e21804, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34383974

RESUMEN

During the last few decades, the consumption of low-calorie sweeteners, as a substitute for caloric sweeteners, has sharply increased. Although research shows that caloric versus low-calorie sweeteners can have differential effects on the brain, it is unknown which neuronal populations are responsible for detecting the difference between the two types of sweeteners. Using in vivo two-photon calcium imaging, we investigated how drinking sucrose or sucralose (a low-calorie sweetener) affects the activity of glutamatergic neurons in the lateral hypothalamus. Furthermore, we explored the consequences of consuming a free-choice high fat diet on the calorie detection abilities of these glutamatergic neurons. We found that glutamatergic neurons indeed can discriminate sucrose from water and sucralose, and that consumption of a free-choice high fat diet shifts the glutamatergic neuronal response from sucrose-specific to sucralose-specific, thereby disrupting calorie detection. These results highlight the disruptive effects of a diet high in saturated fat on calorie detection in the lateral hypothalamus.


Asunto(s)
Ingestión de Energía/fisiología , Área Hipotalámica Lateral/fisiopatología , Animales , Dieta con Restricción de Grasas/métodos , Dieta Alta en Grasa/métodos , Femenino , Área Hipotalámica Lateral/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Edulcorantes/administración & dosificación
4.
Neuropharmacology ; 196: 108702, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34246685

RESUMEN

A growing body of literature implicates noradrenergic (NE) signaling in the modulation of ethanol consumption. However, relatively few studies have detailed specific brain pathways that mediate NE-associated binge-like ethanol consumption. To begin to fill this gap in the literature, male and female C57BL6/J and TH-ires-cre mice underwent pharmacological and chemogenetic testing, respectively, in combination with "drinking in the dark" procedures to model binge-like consumption of ethanol or sucrose solutions. First, we showed that intraperitoneal administration of the NE reuptake inhibitor, reboxetine, blunted binge-like ethanol intake in C57BL6/J mice. Chemogenetic activation of locus coeruleus (LC) tyrosine hydroxylase (TH)-expressing neurons blunted binge-like ethanol intake regardless of sex. Chemogenetic activation of LC projections to the lateral hypothalamus (LH), a region implicated in ethanol consumption, blunted binge-like ethanol drinking without altering sucrose intake in ethanol-experienced or ethanol-naïve mice. In C57BL/6 J mice, LH-targeted microinfusion of an α1-adrenergic receptor (AR) agonist blunted binge-like ethanol intake across both sexes, while LH infusion of a ß-AR agonist blunted binge-like ethanol intake in females exclusively. Finally, in mice with high baseline ethanol intake both an α1- AR agonist and an α-2 AR antagonist blunted binge-like ethanol intake. The present results provide novel evidence that increased NE tone in a circuit arising from the LC and projecting to the LH reduces binge-like ethanol drinking in mice, and may represent a novel approach to treating binge or heavy drinking prior to the development of dependence. This article is part of the special Issue on "Neurocircuitry Modulating Drug and Alcohol Abuse".


Asunto(s)
Inhibidores de Captación Adrenérgica/farmacología , Consumo Excesivo de Bebidas Alcohólicas/metabolismo , Depresores del Sistema Nervioso Central/administración & dosificación , Etanol/administración & dosificación , Área Hipotalámica Lateral/metabolismo , Locus Coeruleus/metabolismo , Norepinefrina/metabolismo , Reboxetina/farmacología , Agonistas de Receptores Adrenérgicos alfa 1/farmacología , Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Agonistas Adrenérgicos beta/farmacología , Animales , Consumo Excesivo de Bebidas Alcohólicas/fisiopatología , Femenino , Área Hipotalámica Lateral/efectos de los fármacos , Área Hipotalámica Lateral/fisiopatología , Locus Coeruleus/efectos de los fármacos , Locus Coeruleus/fisiopatología , Masculino , Ratones , Vías Nerviosas , Tirosina 3-Monooxigenasa
5.
Elife ; 102021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-34042586

RESUMEN

Understanding how neuronal circuits control nociceptive processing will advance the search for novel analgesics. We use functional imaging to demonstrate that lateral hypothalamic parvalbumin-positive (LHPV) glutamatergic neurons respond to acute thermal stimuli and a persistent inflammatory irritant. Moreover, their chemogenetic modulation alters both pain-related behavioral adaptations and the unpleasantness of a noxious stimulus. In two models of persistent pain, optogenetic activation of LHPV neurons or their ventrolateral periaqueductal gray area (vlPAG) axonal projections attenuates nociception, and neuroanatomical tracing reveals that LHPV neurons preferentially target glutamatergic over GABAergic neurons in the vlPAG. By contrast, LHPV projections to the lateral habenula regulate aversion but not nociception. Finally, we find that LHPV activation evokes additive to synergistic antinociceptive interactions with morphine and restores morphine antinociception following the development of morphine tolerance. Our findings identify LHPV neurons as a lateral hypothalamic cell type involved in nociception and demonstrate their potential as a target for analgesia.


Asunto(s)
Conducta Animal , Área Hipotalámica Lateral/fisiopatología , Nocicepción , Dolor/fisiopatología , Dolor/psicología , Analgésicos Opioides/uso terapéutico , Animales , Animales Modificados Genéticamente , Conducta Animal/efectos de los fármacos , Señalización del Calcio , Modelos Animales de Enfermedad , Tolerancia a Medicamentos , Femenino , Neuronas GABAérgicas/metabolismo , Ácido Glutámico/metabolismo , Área Hipotalámica Lateral/efectos de los fármacos , Área Hipotalámica Lateral/metabolismo , Masculino , Ratones Endogámicos C57BL , Microscopía Fluorescente , Morfina/farmacología , Vías Nerviosas/metabolismo , Vías Nerviosas/fisiopatología , Técnicas de Trazados de Vías Neuroanatómicas , Nocicepción/efectos de los fármacos , Optogenética , Dolor/metabolismo , Dolor/prevención & control , Parvalbúminas/genética , Parvalbúminas/metabolismo
6.
Neuroreport ; 32(7): 588-595, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33850090

RESUMEN

OBJECTIVE: Diffuse axonal injury (DAI) is a common pathological process after traumatic brain injury, which may cause survivors severe functional disorders, including cognitive impairment and physical disability. Recent literature indicated lateral hypothalamus and medial hypothalamus damage during DAI. Thus, we aim to investigate whether there is imaging evidence of hypothalamic injury in patients with DAI and its clinical association. METHODS: Twenty-four patients with diagnosed DAI and 26 age and sex-matched healthy controls underwent resting-state functional MRI. We assessed the lateral hypothalamus and medial hypothalamus functional connectivity with seed-based analysis in DAI. Furthermore, a partial correlation was used to measure its clinical association. The prediction of the severity of DAI from the altered lateral hypothalamus and medial hypothalamus connectivity was conducted using a general linear model. RESULTS: Compared with healthy control, the DAI group showed significantly decreased lateral hypothalamus functional connectivity with the basal ganglia and cingulate gyrus, which was positively correlated with mini-mental state examination scores (Bonferroni correction at P < 0.0125). Importantly, this disrupted functional connectivity can be used to predict the patients' cognitive state reliably (P = 0.006; P = 0.009, respectively) in DAI. Moreover, we also observed increased connectivity of medial hypothalamus with the superior temporal gyrus and the regions around the operculum. Furthermore, there was a trend of negative correlation between the medial hypothalamus functional connectivity changes to the right superior temporal gyrus and the disability rating scale scores in the DAI group. CONCLUSION: Our results suggest that there are alterations of medial hypothalamus and lateral hypothalamus connectivity in DAI and further understand its clinical symptoms, including related cognitive impairment.


Asunto(s)
Disfunción Cognitiva/etiología , Lesión Axonal Difusa/complicaciones , Área Hipotalámica Lateral/fisiopatología , Plasticidad Neuronal/fisiología , Adolescente , Adulto , Mapeo Encefálico , Disfunción Cognitiva/diagnóstico por imagen , Disfunción Cognitiva/fisiopatología , Lesión Axonal Difusa/diagnóstico por imagen , Lesión Axonal Difusa/fisiopatología , Femenino , Humanos , Área Hipotalámica Lateral/diagnóstico por imagen , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Adulto Joven
7.
Sci Rep ; 11(1): 5580, 2021 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-33692388

RESUMEN

Deep brain stimulation (DBS) is a powerful neurostimulation therapy proposed for the treatment of several neuropsychiatric disorders. However, DBS mechanism of action remains unclear, being its effects on brain dynamics of particular interest. Specifically, DBS reversibility is a major point of debate. Preclinical studies in obesity showed that the stimulation of the lateral hypothalamus (LH) and nucleus accumbens (NAcc), brain centers involved in satiety and reward circuits, are able to modulate the activity of brain structures impaired in this pathology. Nevertheless, the long-term persistence of this modulation after DBS withdrawal was unexplored. Here we examine the in vivo presence of such changes 1 month after LH- and NAcc-DBS, along with differences in synaptic plasticity, following an exploratory approach. Thus, both stimulated and non-stimulated animals with electrodes in the NAcc showed a common pattern of brain metabolism modulation, presumably derived from the electrodes' presence. In contrast, animals stimulated in the LH showed a relative metabolic invariance, and a reduction of neuroplasticity molecules, evidencing long-lasting neural changes. Our findings suggest that the reversibility or persistence of DBS modulation in the long-term depends on the selected DBS target. Therefore, the DBS footprint would be influenced by the stability achieved in the neural network involved during the stimulation.


Asunto(s)
Estimulación Encefálica Profunda , Área Hipotalámica Lateral/metabolismo , Plasticidad Neuronal , Núcleo Accumbens/metabolismo , Obesidad/metabolismo , Animales , Modelos Animales de Enfermedad , Área Hipotalámica Lateral/fisiopatología , Masculino , Núcleo Accumbens/fisiopatología , Obesidad/fisiopatología , Ratas , Ratas Zucker
8.
Proc Natl Acad Sci U S A ; 117(50): 32155-32164, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33257584

RESUMEN

Anxiety commonly co-occurs with obsessive-compulsive disorder (OCD). Both of them are closely related to stress. However, the shared neurobiological substrates and therapeutic targets remain unclear. Here we report an amelioration of both anxiety and OCD via the histamine presynaptic H3 heteroreceptor on glutamatergic afferent terminals from the prelimbic prefrontal cortex (PrL) to the nucleus accumbens (NAc) core, a vital node in the limbic loop. The NAc core receives direct hypothalamic histaminergic projections, and optogenetic activation of hypothalamic NAc core histaminergic afferents selectively suppresses glutamatergic rather than GABAergic synaptic transmission in the NAc core via the H3 receptor and thus produces an anxiolytic effect and improves anxiety- and obsessive-compulsive-like behaviors induced by restraint stress. Although the H3 receptor is expressed in glutamatergic afferent terminals from the PrL, basolateral amygdala (BLA), and ventral hippocampus (vHipp), rather than the thalamus, only the PrL- and not BLA- and vHipp-NAc core glutamatergic pathways among the glutamatergic afferent inputs to the NAc core is responsible for co-occurrence of anxiety- and obsessive-compulsive-like behaviors. Furthermore, activation of the H3 receptor ameliorates anxiety and obsessive-compulsive-like behaviors induced by optogenetic excitation of the PrL-NAc glutamatergic afferents. These results demonstrate a common mechanism regulating anxiety- and obsessive-compulsive-like behaviors and provide insight into the clinical treatment strategy for OCD with comorbid anxiety by targeting the histamine H3 receptor in the NAc core.


Asunto(s)
Trastornos de Ansiedad/tratamiento farmacológico , Agonistas de los Receptores Histamínicos/administración & dosificación , Núcleo Accumbens/fisiopatología , Trastorno Obsesivo Compulsivo/tratamiento farmacológico , Receptores Histamínicos H3/metabolismo , Vías Aferentes/efectos de los fármacos , Vías Aferentes/fisiopatología , Animales , Trastornos de Ansiedad/etiología , Trastornos de Ansiedad/fisiopatología , Trastornos de Ansiedad/psicología , Modelos Animales de Enfermedad , Glutamatos/metabolismo , Histamina/metabolismo , Antagonistas de los Receptores Histamínicos H3/administración & dosificación , Humanos , Área Hipotalámica Lateral/efectos de los fármacos , Área Hipotalámica Lateral/fisiopatología , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Núcleo Accumbens/citología , Núcleo Accumbens/efectos de los fármacos , Trastorno Obsesivo Compulsivo/etiología , Trastorno Obsesivo Compulsivo/fisiopatología , Trastorno Obsesivo Compulsivo/psicología , Optogenética , Técnicas de Placa-Clamp , Corteza Prefrontal/citología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiopatología , Terminales Presinápticos/efectos de los fármacos , Terminales Presinápticos/metabolismo , Ratas , Ratas Transgénicas , Técnicas Estereotáxicas , Estrés Psicológico/complicaciones , Estrés Psicológico/psicología , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
9.
JCI Insight ; 5(12)2020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32365348

RESUMEN

Imprinted genes are highly expressed in the hypothalamus; however, whether specific imprinted genes affect hypothalamic neuromodulators and their functions is unknown. It has been suggested that Prader-Willi syndrome (PWS), a neurodevelopmental disorder caused by lack of paternal expression at chromosome 15q11-q13, is characterized by hypothalamic insufficiency. Here, we investigate the role of the paternally expressed Snord116 gene within the context of sleep and metabolic abnormalities of PWS, and we report a significant role of this imprinted gene in the function and organization of the 2 main neuromodulatory systems of the lateral hypothalamus (LH) - namely, the orexin (OX) and melanin concentrating hormone (MCH) - systems. We observed that the dynamics between neuronal discharge in the LH and the sleep-wake states of mice with paternal deletion of Snord116 (PWScrm+/p-) are compromised. This abnormal state-dependent neuronal activity is paralleled by a significant reduction in OX neurons in the LH of mutant mice. Therefore, we propose that an imbalance between OX- and MCH-expressing neurons in the LH of mutant mice reflects a series of deficits manifested in the PWS, such as dysregulation of rapid eye movement (REM) sleep, food intake, and temperature control.


Asunto(s)
Conducta Animal/fisiología , Área Hipotalámica Lateral/metabolismo , Hipotálamo/metabolismo , Orexinas/metabolismo , ARN Nucleolar Pequeño/genética , Sueño/fisiología , Animales , Modelos Animales de Enfermedad , Conducta Alimentaria , Área Hipotalámica Lateral/fisiopatología , Hormonas Hipotalámicas/metabolismo , Melaninas/metabolismo , Ratones , Neuronas/metabolismo , Hormonas Hipofisarias/metabolismo , Síndrome de Prader-Willi/metabolismo , Síndrome de Prader-Willi/fisiopatología
10.
Neuron ; 107(1): 158-172.e4, 2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32333845

RESUMEN

Overeating typically follows periods of energy deficit, but it is also sustained by highly palatable foods, even without metabolic demand. Dopamine D1 receptor-expressing medium spiny neurons (D1-MSNs) of the nucleus accumbens shell (NAcSh) project to the lateral hypothalamus (LH) to authorize feeding when inhibited. Whether plasticity at these synapses can affect food intake is unknown. Here, ex vivo electrophysiology recordings reveal that D1-MSN-to-LH inhibitory transmission is depressed in circumstances in which overeating is promoted. Endocannabinoid signaling is identified as the induction mechanism, since inhibitory plasticity and concomitant overeating were blocked or induced by CB1R antagonism or agonism, respectively. D1-MSN-to-LH projectors were largely non-overlapping with D1-MSNs targeting ventral pallidum or ventral midbrain, providing an anatomical basis for distinct circuit plasticity mechanisms. Our study reveals a critical role for plasticity at D1-MSN-to-LH synapses in adaptive feeding control, which may underlie persistent overeating of unhealthy foods, a major risk factor for developing obesity.


Asunto(s)
Hiperfagia/fisiopatología , Área Hipotalámica Lateral/fisiopatología , Depresión Sináptica a Largo Plazo/fisiología , Núcleo Accumbens/fisiopatología , Transmisión Sináptica/fisiología , Animales , Ratones , Vías Nerviosas/fisiopatología
11.
Brain Res ; 1732: 146674, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31981680

RESUMEN

To date several circuities in supraspinal site of the central nervous system have been known to engage in pain modulation. Lateral hypothalamus (LH) is known as part of the circuit of pain modulation among supraspinal sites. Its role in several animal pain models has been well defined. In this study, we examined the role of spinal orexin receptors in antinociceptive response elicited by the LH stimulation in an animal model of neuropathic pain. Male Wistar rats were unilaterally implanted with a cannula into the LH and a catheter into the L4-L5 segments of the spinal cord followed by chronic constriction injury (CCI) surgery. Intra-LH microinjection of carbachol (500 nM; 0.5 µL) was done 5 min after intrathecal administration of the orexin receptor antagonists, SB-334867 or TCS OX2 29; control animals received DMSO. Mechanical allodynia and thermal hyperalgesia were evaluated using von Frey filaments and a thermal stimulus. The results showed that carbachol induces antiallodynic and anti-thermal hyperalgesic effects in a dose-dependent manner. The antiallodynic and anti-thermal hyperalgesic effects induced by intra-LH injection of carbachol were reversed by intrathecal administration of 10 µL-100 nM solutions of SB-334867 or TCS OX2 in neuropathic rats. However, solely intrathecal administration of both antagonists had no effect in neuropathic rats. There appears to be a neural pathway from the LH to the spinal cord, which potentially contributes to the modulation of neuropathic pain. The implications are that there may be novel therapeutic approaches for the treatment of people suffered from chronic neuropathic pain in clinic.


Asunto(s)
Hiperalgesia/metabolismo , Área Hipotalámica Lateral/efectos de los fármacos , Neuralgia/metabolismo , Receptores de Orexina/metabolismo , Umbral del Dolor/efectos de los fármacos , Médula Espinal/metabolismo , Animales , Benzoxazoles/farmacología , Carbacol/farmacología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Calor , Hiperalgesia/fisiopatología , Área Hipotalámica Lateral/metabolismo , Área Hipotalámica Lateral/fisiopatología , Isoquinolinas/farmacología , Naftiridinas/farmacología , Neuralgia/fisiopatología , Antagonistas de los Receptores de Orexina/farmacología , Manejo del Dolor , Piridinas/farmacología , Ratas , Ratas Wistar , Médula Espinal/fisiopatología , Urea/análogos & derivados , Urea/farmacología
12.
Neuroendocrinology ; 110(6): 552-562, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31484186

RESUMEN

The hypothalamus contains integrative systems that support life, including physiological processes such as food intake, energy expenditure, and reproduction. Here, we show that anorexia nervosa (AN) patients, contrary to normal weight and constitutionally lean individuals, respond with a paradoxical reduction in hypothalamic levels of glutamate/glutamine (Glx) upon feeding. This reversal of the Glx response is associated with decreased wiring in the arcuate nucleus and increased connectivity in the lateral hypothalamic area, which are involved in the regulation on a variety of physiological and behavioral functions including the control of food intake and energy balance. The identification of distinct hypothalamic neurochemical dysfunctions and associated structural variations in AN paves the way for the development of new diagnostic and treatment strategies in conditions associated with abnormal body mass index and a maladaptive response to negative energy balance.


Asunto(s)
Anorexia Nerviosa , Núcleo Arqueado del Hipotálamo , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Área Hipotalámica Lateral , Adulto , Anorexia Nerviosa/diagnóstico por imagen , Anorexia Nerviosa/metabolismo , Anorexia Nerviosa/patología , Anorexia Nerviosa/fisiopatología , Núcleo Arqueado del Hipotálamo/diagnóstico por imagen , Núcleo Arqueado del Hipotálamo/metabolismo , Núcleo Arqueado del Hipotálamo/patología , Núcleo Arqueado del Hipotálamo/fisiopatología , Femenino , Humanos , Área Hipotalámica Lateral/diagnóstico por imagen , Área Hipotalámica Lateral/metabolismo , Área Hipotalámica Lateral/patología , Área Hipotalámica Lateral/fisiopatología , Imagen por Resonancia Magnética , Masculino , Espectroscopía de Protones por Resonancia Magnética , Adulto Joven
13.
Science ; 364(6447): 1271-1274, 2019 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-31249056

RESUMEN

The current obesity epidemic is a major worldwide health concern. Despite the consensus that the brain regulates energy homeostasis, the neural adaptations governing obesity are unknown. Using a combination of high-throughput single-cell RNA sequencing and longitudinal in vivo two-photon calcium imaging, we surveyed functional alterations of the lateral hypothalamic area (LHA)-a highly conserved brain region that orchestrates feeding-in a mouse model of obesity. The transcriptional profile of LHA glutamatergic neurons was affected by obesity, exhibiting changes indicative of altered neuronal activity. Encoding properties of individual LHA glutamatergic neurons were then tracked throughout obesity, revealing greatly attenuated reward responses. These data demonstrate how diet disrupts the function of an endogenous feeding suppression system to promote overeating and obesity.


Asunto(s)
Área Hipotalámica Lateral/metabolismo , Área Hipotalámica Lateral/fisiopatología , Obesidad/genética , Obesidad/fisiopatología , Transcriptoma , Animales , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Ácido Glutámico/metabolismo , Ratones , Neuronas , Obesidad/psicología , Recompensa , Proteína 2 de Transporte Vesicular de Glutamato/genética
14.
Neurobiol Dis ; 109(Pt A): 54-63, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29017795

RESUMEN

Lack of adequate sleep has become increasingly common in our 24/7 society. Unfortunately diminished sleep has significant health consequences including metabolic and cardiovascular disease and mental disorders including depression. The pathways by which reduced sleep adversely affects physiology and behavior are unknown. We found that 6h of sleep deprivation in adult male rats induces changes in neuronal activity in the lateral hypothalamus, the paraventricular nucleus, the arcuate nucleus and the mammillary bodies. Surprisingly, these alterations last for up to 48h. The data show that sleep loss has prolonged effects on the activity of multiple hypothalamic areas. Our data indicate also that measuring electroencephalographic slow wave activity underestimates the amount of time that the hypothalamus requires to recover from episodes of sleep deprivation. We propose that these hypothalamic changes underlie the well-established relationship between sleep loss and several diseases such as metabolic disorders, stress and depression and that sufficient sleep is vital for autonomic functions controlled by the hypothalamus.


Asunto(s)
Hipotálamo/fisiopatología , Neuronas/fisiología , Privación de Sueño/fisiopatología , Animales , Núcleo Arqueado del Hipotálamo/fisiopatología , Electroencefalografía , Área Hipotalámica Lateral/fisiopatología , Masculino , Tubérculos Mamilares/fisiopatología , Núcleo Hipotalámico Paraventricular/fisiopatología , Ratas Wistar
15.
PLoS One ; 12(5): e0177551, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28542167

RESUMEN

Changes in the hypothalamic-pituitary-adrenal (HPA) axis activity constitute a key component of bipolar mania, but the extent and nature of these alterations are not fully understood. We use here the lateral hypothalamic-kindled (LHK) rat model to deliberately induce an acute manic-like episode and measure serum corticosterone concentrations to assess changes in HPA axis activity. A mathematical model is developed to succinctly describe the entwined biochemical transformations that underlay the HPA axis and emulate by numerical simulations the considerable increase in serum corticosterone concentration induced by LHK. Synergistic combination of the LHK rat model and dynamical systems theory allows us to quantitatively characterize changes in HPA axis activity under controlled induction of acute manic-like states and provides a framework to study in silico how the dynamic integration of neurochemical transformations underlying the HPA axis is disrupted in these states.


Asunto(s)
Trastorno Bipolar/sangre , Trastorno Bipolar/fisiopatología , Corticosterona/sangre , Área Hipotalámica Lateral/fisiopatología , Modelos Biológicos , Animales , Masculino , Sistema Hipófiso-Suprarrenal/fisiopatología , Ratas , Ratas Wistar
16.
Behav Brain Res ; 328: 195-208, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28432009

RESUMEN

The lateral habenula (LHb) is an epithalamic brain region implicated in aversive processing via negative modulation of midbrain dopamine (DA) and serotonin (5-HT) systems. Given the role of the LHb in inhibiting DA and 5-HT systems, it is thought to be involved in various psychiatric pathologies, including drug addiction. In support, it has been shown that LHb plays a critical role in cocaine- and ethanol-related behaviors, most likely by mediating drug-induced aversive conditioning. In our previous work, we showed that LHb lesions increased voluntary ethanol consumption and operant ethanol self-administration and blocked yohimbine-induced reinstatement of ethanol self-administration. LHb lesions also attenuated ethanol-induced conditioned taste aversion suggesting that a mechanism for the increased intake of ethanol may be reduced aversion learning. However, whether afferents to the LHb are required for mediating effects of the LHb on these behaviors remained to be investigated. Our present results show that lesioning the fiber bundle carrying afferent inputs to the LHb, the stria medullaris (SM), increases voluntary ethanol consumption, suggesting that afferent structures projecting to the LHb are important for mediating ethanol-directed behaviors. We then chose two afferent structures as the focus of our investigation. We specifically studied the role of the inputs from the lateral hypothalamus (LH) and ventral pallidum (VP) to the LHb in ethanol-directed behaviors. Our results show that the LH-LHb projection is necessary for regulating voluntary ethanol consumption. These results are an important first step towards understanding the functional role of afferents to LHb with regard to ethanol consumption.


Asunto(s)
Consumo de Bebidas Alcohólicas/fisiopatología , Prosencéfalo Basal/fisiopatología , Habénula/fisiopatología , Área Hipotalámica Lateral/fisiopatología , Consumo de Bebidas Alcohólicas/patología , Animales , Prosencéfalo Basal/patología , Depresores del Sistema Nervioso Central/administración & dosificación , Comportamiento de Búsqueda de Drogas/fisiología , Etanol/administración & dosificación , Habénula/patología , Área Hipotalámica Lateral/patología , Masculino , Vías Nerviosas/patología , Vías Nerviosas/fisiopatología , Ratas Long-Evans , Autoadministración , Volición
17.
Metabolism ; 67: 14-25, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28081774

RESUMEN

Nesfatin-1, a recently discovered neuropeptide involved in satiety. Recent studies have revealed that central nesfatin-1 inhibits gastric emptying and gastric acid secretion, though the mechanisms involved in these processes are not known. We aim to explore the effects of nesfatin-1 on a population of gastric distension (GD)-sensitive neurons in the lateral hypothalamus (LHA), gastric motility, and gastric secretion and the role for an arcuate nucleus (Arc)-LHA neural pathway in these processes. Single unit extracellular discharge recordings were made in of LHA. Further, gastric motility and gastric secretion in rats were monitored. Retrograde tracing and fluorescent immunohistochemical staining were used to explore nesfatin-1 neuron projection. The results revealed that administration of nesfatin-1 to the LHA or electric stimulation of the Arc could alter the neuronal activity of melanin-concentrating hormone (MCH)-responsive, GD-responsive neurons in LHA, which could be blocked by pretreatment with MCH receptor-1 antagonist PMC-3881-PI or weakened by pretreatment of a nesfatin-1 antibody in LHA. Administration of nesfatin-1 into LHA could inhibit gastric motility and gastric secretion, and these effects could be enhanced by administration of PMC-3881-PI. Electrical stimulation of Arc promoted the gastric motility and gastric secretion. Nesfatin-1 antibody or PMC-3881-PI pretreatment to LHA had no effect on Arc stimulation-induced gastric motility, but these pretreatments did alter Arc stimulation-induced effects on gastric secretion. Our findings suggest that nesfatin-1 signaling in LHA participates in the regulation of efferent information from the gastrointestinal tract and gastric secretion which also involve MCH signaling. Further, they show that a nesfatin-1-positive Arc to LHA pathway is critical for these effects.


Asunto(s)
Núcleo Arqueado del Hipotálamo/fisiopatología , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al ADN/metabolismo , Dilatación Gástrica/metabolismo , Área Hipotalámica Lateral/metabolismo , Hormonas Hipotalámicas/farmacología , Melaninas/farmacología , Proteínas del Tejido Nervioso/metabolismo , Hormonas Hipofisarias/farmacología , Animales , Anticuerpos Bloqueadores/farmacología , Proteínas de Unión al Calcio/antagonistas & inhibidores , Proteínas de Unión al ADN/antagonistas & inhibidores , Estimulación Eléctrica , Dilatación Gástrica/fisiopatología , Motilidad Gastrointestinal , Área Hipotalámica Lateral/fisiopatología , Masculino , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Neuronas , Nucleobindinas , Oligopéptidos/farmacología , Ratas , Ratas Wistar , Estómago/inervación
18.
Neuroscience ; 324: 82-91, 2016 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-26964685

RESUMEN

In homeotherms, the hypothalamus controls thermoregulatory and adaptive mechanisms in energy balance, sleep-wake and locomotor activity to maintain optimal body temperature. Orexin neurons may be involved in these functions as they promote thermogenesis, food intake and behavioral arousal, and are sensitive to temperature and metabolic status. How thermal and energy balance signals are integrated in these neurons is unknown. Thus, we investigated the cellular mechanisms of thermosensing in orexin neurons and their response to a change in energy status using whole-cell patch clamp on rat brain slices. We found that warming induced an increase in miniature excitatory postsynaptic current (EPSC) frequency, which was blocked by the transient receptor potential vanilloid-1 (TRPV1) receptor antagonist AMG9810 and mimicked by its agonist capsaicin, suggesting that the synaptic effect is mediated by heat-sensitive TRPV1 channels. Furthermore, warming inhibits orexin neurons by activating ATP-sensitive potassium (KATP) channels, an effect regulated by uncoupling protein 2 (UCP2), as the UCP2 inhibitor genipin abolished this response. These properties are unique to orexin neurons in the lateral hypothalamus, as neighboring melanin-concentrating hormone neurons showed no response to warming within the physiological temperature range. Interestingly, in rats fed with western diet for 1 or 11weeks, orexin neurons had impaired synaptic and KATP response to warming. In summary, this study reveals several mechanisms underlying thermosensing in orexin neurons and their attenuation by western diet. Overeating induced by western diet may in part be due to impaired orexin thermosensing, as post-prandial thermogenesis may promote satiety and lethargy by inhibiting orexin neurons.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Calor , Neuronas/fisiología , Orexinas/metabolismo , Sensación Térmica/fisiología , Acrilamidas/farmacología , Animales , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Capsaicina/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Área Hipotalámica Lateral/efectos de los fármacos , Área Hipotalámica Lateral/fisiopatología , Hormonas Hipotalámicas/metabolismo , Iridoides/farmacología , Canales KATP/metabolismo , Masculino , Melaninas/metabolismo , Potenciales Postsinápticos Miniatura/efectos de los fármacos , Potenciales Postsinápticos Miniatura/fisiología , Neuronas/efectos de los fármacos , Neurotransmisores/farmacología , Técnicas de Placa-Clamp , Hormonas Hipofisarias/metabolismo , Ratas Sprague-Dawley , Canales Catiónicos TRPV/agonistas , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/metabolismo , Sensación Térmica/efectos de los fármacos , Técnicas de Cultivo de Tejidos , Proteína Desacopladora 2/antagonistas & inhibidores , Proteína Desacopladora 2/metabolismo
19.
Behav Brain Res ; 298(Pt B): 167-80, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26554726

RESUMEN

The parvafox nucleus is located ventrolaterally in the lateral hypothalamic area (LHA). Its core and shell are composed of neurons expressing the calcium-binding protein parvalbumin (PV) and the transcription factor Foxb1, respectively. Given the known functions of the LHA and that the parvafox nucleus receives afferents from the lateral orbitofrontal cortex and projects to the periaqueductal gray matter, a functional role of this entity in the expression of positive emotions has been postulated. The purpose of the present study was to ascertain whether the deletion of neurons in the parvafox nucleus influenced the tickling-induced 50-kHz calls, which are thought to reflect positive affective states, in rats. To this end, tickling of the animals (heterospecific play) was combined with intracerebral injections of the excitotoxin kainic acid into the parvafox nucleus. The most pronounced surgery-associated reduction in 50-kHz call-numbers was observed in the group of rats in which, on the basis of PV-immunoreactive-cell counts in the parvafox nucleus, bilateral lesions had been successfully produced. Two other parameters that were implemented to quantify positive affective behaviour, namely, an approach towards and a following of the hand of the tickling experimenter, were likewise most markedly suppressed in the group of rats with bilaterally successful lesions. Furthermore, positive correlations were found between each of the investigated parameters. Our data afford evidence that the parvafox nucleus plays a role in the production of 50-kHz calls in rats, and, more generally, in the expression of positive emotions.


Asunto(s)
Afecto/fisiología , Conducta de Elección/fisiología , Área Hipotalámica Lateral/fisiología , Conducta Social , Percepción del Tacto/fisiología , Vocalización Animal/fisiología , Animales , Recuento de Células , Estudios de Cohortes , Femenino , Área Hipotalámica Lateral/fisiopatología , Ácido Kaínico , Masculino , Microglía/fisiología , Actividad Motora/fisiología , Parvalbúminas/metabolismo , Ratas Wistar , Ultrasonido
20.
Neuroscience ; 315: 1-17, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26691962

RESUMEN

Syrian hamsters exposed to anabolic/androgenic steroids (AAS) during adolescence consistently show increased aggressive behavior across studies. Although the behavioral and anatomical profiles of AAS-induced alterations have been well characterized, there is a lack of data describing physiological changes that accompany these alterations. For instance, behavioral pharmacology and neuroanatomical studies show that AAS-induced changes in the vasopressin (AVP) neural system within the latero-anterior hypothalamus (LAH) interact with the serotonin (5HT) and dopamine (DA) systems to modulate aggression. To characterize the electrophysiological profile of the AAS aggression circuit, we recorded LAH neurons in adolescent male hamsters in vivo and microiontophoretically applied agonists and antagonists of aggressive behavior. The interspike interval (ISI) of neurons from AAS-treated animals correlated positively with aggressive behaviors, and adolescent AAS exposure altered parameters of activity in regular firing neurons while also changing the proportion of neuron types (i.e., bursting, regular, irregular). AAS-treated animals had more responsive neurons that were excited by AVP application, while cells from control animals showed the opposite effect and were predominantly inhibited by AVP. Both DA D2 antagonists and 5HT increased the firing frequency of AVP-responsive cells from AAS animals and dual application of AVP and D2 antagonists doubled the excitatory effect of AVP or D2 antagonist administration alone. These data suggest that multiple DA circuits in the LAH modulate AAS-induced aggressive responding. More broadly, these data show that multiple neurochemical interactions at the neurophysiological level are altered by adolescent AAS exposure.


Asunto(s)
Agresión/efectos de los fármacos , Anabolizantes/toxicidad , Área Hipotalámica Lateral/efectos de los fármacos , Hipotálamo Anterior/efectos de los fármacos , Neuronas/efectos de los fármacos , Esteroides/toxicidad , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Agresión/fisiología , Animales , Antagonistas de los Receptores de Dopamina D2/farmacología , Área Hipotalámica Lateral/fisiopatología , Hipotálamo Anterior/fisiopatología , Masculino , Mesocricetus , Neuronas/fisiología , Receptores de Dopamina D2/metabolismo , Salicilamidas/farmacología , Serotonina/administración & dosificación , Serotonina/metabolismo , Vasopresinas/administración & dosificación , Vasopresinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...