Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sleep ; 46(9)2023 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-37224457

RESUMEN

A workshop titled "Beyond the Symptom: The Biology of Fatigue" was held virtually September 27-28, 2021. It was jointly organized by the Sleep Research Society and the Neurobiology of Fatigue Working Group of the NIH Blueprint Neuroscience Research Program. For access to the presentations and video recordings, see: https://neuroscienceblueprint.nih.gov/about/event/beyond-symptom-biology-fatigue. The goals of this workshop were to bring together clinicians and scientists who use a variety of research approaches to understand fatigue in multiple conditions and to identify key gaps in our understanding of the biology of fatigue. This workshop summary distills key issues discussed in this workshop and provides a list of promising directions for future research on this topic. We do not attempt to provide a comprehensive review of the state of our understanding of fatigue, nor to provide a comprehensive reprise of the many excellent presentations. Rather, our goal is to highlight key advances and to focus on questions and future approaches to answering them.


Asunto(s)
Fatiga , Motivación , Humanos , Biología
2.
Nat Commun ; 13(1): 4163, 2022 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-35851580

RESUMEN

Humans and animals lacking orexin neurons exhibit daytime sleepiness, sleep attacks, and state instability. While the circuit basis by which orexin neurons contribute to consolidated wakefulness remains unclear, existing models posit that orexin neurons provide their wake-stabilizing influence by exerting excitatory tone on other brain arousal nodes. Here we show using in vivo optogenetics, in vitro optogenetic-based circuit mapping, and single-cell transcriptomics that orexin neurons also contribute to arousal maintenance through indirect inhibition of sleep-promoting neurons of the ventrolateral preoptic nucleus. Activation of this subcortical circuit rapidly drives wakefulness from sleep by differentially modulating the activity of ventrolateral preoptic neurons. We further identify and characterize a feedforward circuit through which orexin (and co-released glutamate) acts to indirectly target and inhibit sleep-promoting ventrolateral preoptic neurons to produce arousal. This revealed circuitry provides an alternate framework for understanding how orexin neurons contribute to the maintenance of consolidated wakefulness and stabilize behavioral state.


Asunto(s)
Nivel de Alerta , Sueño , Animales , Nivel de Alerta/fisiología , Humanos , Neuronas/fisiología , Orexinas , Sueño/fisiología , Vigilia/fisiología
3.
Science ; 375(6584): 972-973, 2022 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-35239395

RESUMEN

How does dopamine, the brain's pleasure signal, regulate the dream stage of sleep?


Asunto(s)
Sueños , Sueño REM , Dopamina , Sueños/fisiología , Sueño , Sueño REM/fisiología
4.
Int J Mol Sci ; 23(6)2022 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-35328326

RESUMEN

For over a century, the role of the preoptic hypothalamus and adjacent basal forebrain in sleep-wake regulation has been recognized. However, for years, the identity and location of sleep- and wake-promoting neurons in this region remained largely unresolved. Twenty-five years ago, Saper and colleagues uncovered a small collection of sleep-active neurons in the ventrolateral preoptic nucleus (VLPO) of the preoptic hypothalamus, and since this seminal discovery the VLPO has been intensively investigated by labs around the world, including our own. Herein, we first review the history of the preoptic area, with an emphasis on the VLPO in sleep-wake control. We then attempt to synthesize our current understanding of the circuit, cellular and synaptic bases by which the VLPO both regulates and is itself regulated, in order to exert a powerful control over behavioral state, as well as examining data suggesting an involvement of the VLPO in other physiological processes.


Asunto(s)
Área Preóptica , Sueño , Hipotálamo , Aprendizaje , Neuronas/fisiología , Sueño/fisiología
5.
Curr Top Behav Neurosci ; 59: 447-468, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34595740

RESUMEN

Histamine is a small monoamine signaling molecule that plays a role in many peripheral and central physiological processes, including the regulation of wakefulness. The tuberomammillary nucleus is the sole neuronal source of histamine in the brain, and histamine neurons are thought to promote wakefulness and vigilance maintenance - under certain environmental and/or behavioral contexts - through their diffuse innervation of the cortex and other wake-promoting brain circuits. Histamine neurons also contain a number of other putative neurotransmitters, although the functional role of these co-transmitters remains incompletely understood. Within the brain histamine operates through three receptor subtypes that are located on pre- and post-synaptic membranes. Some histamine receptors exhibit constitutive activity, and hence exist in an activated state even in the absence of histamine. Newer medications used to reduce sleepiness in narcolepsy patients in fact enhance histamine signaling by blunting the constitutive activity of these histamine receptors. In this chapter, we provide an overview of the central histamine system with an emphasis on its role in behavioral state regulation and how drugs targeting histamine receptors are used clinically to treat a wide range of sleep-wake disorders.


Asunto(s)
Histamina , Sueño , Histamina/fisiología , Humanos , Área Hipotalámica Lateral/fisiología , Receptores Histamínicos , Sueño/fisiología , Vigilia/fisiología
6.
Diabetologia ; 64(11): 2575-2588, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34430981

RESUMEN

AIMS/HYPOTHESIS: Hypothalamic inflammation and sympathetic nervous system hyperactivity are hallmark features of the metabolic syndrome and type 2 diabetes. Hypothalamic inflammation may aggravate metabolic and immunological pathologies due to extensive sympathetic activation of peripheral tissues. Loss of somatostatinergic (SST) neurons may contribute to enhanced hypothalamic inflammation. METHODS: The present data show that leptin receptor-deficient (db/db) mice exhibit reduced hypothalamic SST neurons, particularly in the periventricular nucleus. We model this finding, using adeno-associated virus delivery of diphtheria toxin subunit A (DTA) driven by an SST-cre system to deplete these neurons in Sstcre/gfp mice (SST-DTA). RESULTS: SST-DTA mice exhibit enhanced hypothalamic c-Fos expression and brain inflammation as demonstrated by microglial and astrocytic activation. Bone marrow from SST-DTA mice undergoes skewed haematopoiesis, generating excess granulocyte-monocyte progenitors and increased proinflammatory (C-C chemokine receptor type 2; CCR2hi) monocytes. SST-DTA mice exhibited a 'diabetic retinopathy-like' phenotype: reduced visual function by optokinetic response (0.4 vs 0.25 cycles/degree; SST-DTA vs control mice); delayed electroretinogram oscillatory potentials; and increased percentages of retinal monocytes. Finally, mesenteric visceral adipose tissue from SST-DTA mice was resistant to catecholamine-induced lipolysis, displaying 50% reduction in isoprenaline (isoproterenol)-induced lipolysis compared with control littermates. Importantly, hyperglycaemia was not observed in SST-DTA mice. CONCLUSIONS/INTERPRETATION: The isolated reduction in hypothalamic SST neurons was able to recapitulate several hallmark features of type 2 diabetes in disease-relevant tissues.


Asunto(s)
Tejido Adiposo/metabolismo , Médula Ósea/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Hipotálamo/metabolismo , Neuronas/metabolismo , Retina/metabolismo , Somatostatina/metabolismo , Animales , Encéfalo/metabolismo , Diabetes Mellitus Tipo 2/diagnóstico , Toxina Diftérica/toxicidad , Electrorretinografía , Citometría de Flujo , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa
7.
Trends Pharmacol Sci ; 42(5): 329-339, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33781582

RESUMEN

Every cell has a highly sophisticated system for regulating heme levels, which is particularly important with regard to turnover. Heme degradation generates CO and while CO has long been viewed as a metabolic waste product, and at higher concentrations cellularly lethal, we now know that CO is an indispensable gasotransmitter that participates in fundamental physiological processes necessary for survival. Irrefutable preclinical data have resulted in concerted efforts to develop CO as a safe and effective therapeutic agent, but against this notion lies dogma that CO is a poison, especially to the brain. The emergence of this debate is discussed here highlighting the neuroprotective properties of CO through its role on the central circadian clock and ongoing strategies being developed for CO administration for clinical use.


Asunto(s)
Relojes Circadianos , Gasotransmisores , Venenos , Monóxido de Carbono , Hemo Oxigenasa (Desciclizante)
8.
Curr Biol ; 30(23): 4579-4593.e7, 2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-32976803

RESUMEN

Locomotion requires energy, yet animals need to increase locomotion in order to find and consume food in energy-deprived states. While such energy homeostatic coordination suggests brain origin, whether the central melanocortin 4 receptor (Mc4r) system directly modulates locomotion through motor circuits is unknown. Here, we report that hypothalamic Pomc neurons in zebrafish and mice have long-range projections into spinal cord regions harboring Mc4r-expressing V2a interneurons, crucial components of the premotor networks. Furthermore, in zebrafish, Mc4r activation decreases the excitability of spinal V2a neurons as well as swimming and foraging, while systemic or V2a neuron-specific blockage of Mc4r promotes locomotion. In contrast, in mice, electrophysiological recordings revealed that two-thirds of V2a neurons in lamina X are excited by the Mc4r agonist α-MSH, and acute inhibition of Mc4r signaling reduces locomotor activity. In addition, we found other Mc4r neurons in spinal lamina X that are inhibited by α-MSH, which is in line with previous studies in rodents where Mc4r agonists reduced locomotor activity. Collectively, our studies identify spinal V2a interneurons as evolutionary conserved second-order neurons of the central Mc4r system, providing a direct anatomical and functional link between energy homeostasis and locomotor control systems. The net effects of this modulatory system on locomotor activity can vary between different vertebrate species and, possibly, even within one species. We discuss the biological sense of this phenomenon in light of the ambiguity of locomotion on energy balance and the different living conditions of the different species.


Asunto(s)
Núcleo Arqueado del Hipotálamo/fisiología , Interneuronas/metabolismo , Locomoción/fisiología , Proopiomelanocortina/metabolismo , Médula Espinal/fisiología , Proteínas de Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Núcleo Arqueado del Hipotálamo/citología , Evolución Biológica , Fenómenos Electrofisiológicos/efectos de los fármacos , Ratones , Modelos Animales , Red Nerviosa/fisiología , Proopiomelanocortina/genética , Receptor de Melanocortina Tipo 4/agonistas , Receptor de Melanocortina Tipo 4/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Pez Cebra , Proteínas de Pez Cebra/agonistas , Proteínas de Pez Cebra/genética
9.
Nat Commun ; 11(1): 4410, 2020 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-32879310

RESUMEN

The hypothalamic suprachiasmatic (SCN) clock contains several neurochemically defined cell groups that contribute to the genesis of circadian rhythms. Using cell-specific and genetically targeted approaches we have confirmed an indispensable role for vasoactive intestinal polypeptide-expressing SCN (SCNVIP) neurons, including their molecular clock, in generating the mammalian locomotor activity (LMA) circadian rhythm. Optogenetic-assisted circuit mapping revealed functional, di-synaptic connectivity between SCNVIP neurons and dorsomedial hypothalamic neurons, providing a circuit substrate by which SCNVIP neurons may regulate LMA rhythms. In vivo photometry revealed that while SCNVIP neurons are acutely responsive to light, their activity is otherwise behavioral state invariant. Single-nuclei RNA-sequencing revealed that SCNVIP neurons comprise two transcriptionally distinct subtypes, including putative pacemaker and non-pacemaker populations. Altogether, our work establishes necessity of SCNVIP neurons for the LMA circadian rhythm, elucidates organization of circadian outflow from and modulatory input to SCNVIP cells, and demonstrates a subpopulation-level molecular heterogeneity that suggests distinct functions for specific SCNVIP subtypes.


Asunto(s)
Ritmo Circadiano/fisiología , Neuronas/metabolismo , Núcleo Supraquiasmático , Animales , Mapeo Encefálico , Relojes Circadianos/fisiología , Locomoción/fisiología , Ratones , Optogenética/métodos , Núcleo Supraquiasmático/citología , Núcleo Supraquiasmático/metabolismo
10.
BMJ Open ; 10(7): e038474, 2020 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-32690536

RESUMEN

INTRODUCTION: Insomnia frequently occurs in patients admitted to an intensive care unit (ICU). Sleep-promoting agents may reduce rapid eye movement sleep and have deliriogenic effects. Suvorexant (Belsomra) is an orexin receptor antagonist with Food and Drug Administration (FDA) approval for the treatment of adult insomnia, which improves sleep onset and maintenance as well as subjective measures of quality of sleep. This trial will evaluate the efficacy of postoperative oral suvorexant treatment on night-time wakefulness after persistent sleep onset as well as the incidence and duration of delirium among adult cardiac surgical patients. METHODS AND ANALYSIS: In this single-centre, randomised, double-blind, placebo-controlled trial, we will enrol 120 patients, aged 60 years or older, undergoing elective cardiac surgery with planned postoperative admission to the ICU. Participants will be randomised to receive oral suvorexant (20 mg) or placebo one time a day starting the night after extubation. The primary outcome will be wakefulness after persistent sleep onset. The secondary outcome will be total sleep time. Exploratory outcomes will include time to sleep onset, incidence of postoperative in-hospital delirium, number of delirium-free days and subjective sleep quality. ETHICS AND DISSEMINATION: Ethics approval was obtained through the 'Committee on Clinical Investigations' at Beth Israel Deaconess Medical Center (protocol number 2019P000759). The findings will be published in peer-reviewed journals. TRIAL REGISTRATION NUMBER: This trial has been registered at clinicaltrials.gov on 17 September 2019 (NCT04092894).


Asunto(s)
Delirio , Adulto , Azepinas , Delirio/tratamiento farmacológico , Delirio/prevención & control , Método Doble Ciego , Femenino , Humanos , Unidades de Cuidados Intensivos , Persona de Mediana Edad , Antagonistas de los Receptores de Orexina/farmacología , Antagonistas de los Receptores de Orexina/uso terapéutico , Embarazo , Ensayos Clínicos Controlados Aleatorios como Asunto , Sueño/efectos de los fármacos , Volumen Sistólico , Resultado del Tratamiento , Triazoles , Función Ventricular Izquierda
11.
Nat Commun ; 11(1): 2769, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32488015

RESUMEN

During obstructive sleep apnea, elevation of CO2 during apneas contributes to awakening and restoring airway patency. We previously found that glutamatergic neurons in the external lateral parabrachial nucleus (PBel) containing calcitonin gene related peptide (PBelCGRP neurons) are critical for causing arousal during hypercapnia. However, others found that genetic deletion of serotonin (5HT) neurons in the brainstem also prevented arousal from hypercapnia. To examine interactions between the two systems, we showed that dorsal raphe (DR) 5HT neurons selectively targeted the PBel. Either genetically directed deletion or acute optogenetic silencing of DRSert neurons dramatically increased the latency of mice to arouse during hypercapnia, as did silencing DRSert terminals in the PBel. This effect was mediated by 5HT2a receptors which are expressed by PBelCGRP neurons. Our results indicate that the serotonergic input from the DR to the PBel via 5HT2a receptors is critical for modulating the sensitivity of the PBelCGRP neurons that cause arousal to rising levels of blood CO2.


Asunto(s)
Nivel de Alerta/fisiología , Núcleo Dorsal del Rafe/metabolismo , Hipercapnia/metabolismo , Neuronas Serotoninérgicas/metabolismo , Animales , Tronco Encefálico/metabolismo , Péptido Relacionado con Gen de Calcitonina/metabolismo , Dióxido de Carbono , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Transgénicos , Optogenética , Núcleos Parabraquiales , Serotonina/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética
12.
Sleep ; 43(2)2020 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-31553451

RESUMEN

A role for the brain's serotoninergic (5HT) system in the regulation of sleep and wakefulness has been long suggested. Yet, previous studies employing pharmacological, lesion and genetically driven approaches have produced inconsistent findings, leaving 5HT's role in sleep-wake regulation incompletely understood. Here we sought to define the specific contribution of 5HT neurons within the dorsal raphe nucleus (DRN5HT) to sleep and arousal control. To do this, we employed a chemogenetic strategy to selectively and acutely activate DRN5HT neurons and monitored sleep-wake using electroencephalogram recordings. We additionally assessed indices of anxiety using the open field and elevated plus maze behavioral tests and employed telemetric-based recordings to test effects of acute DRN5HT activation on body temperature and locomotor activity. Our findings indicate that the DRN5HT cell population may not modulate sleep-wake per se, but rather that its activation has apparent anxiolytic properties, suggesting the more nuanced view that DRN5HT neurons are sleep permissive under circumstances that produce anxiety or stress.


Asunto(s)
Núcleo Dorsal del Rafe , Sueño , Nivel de Alerta , Neuronas Serotoninérgicas , Vigilia
13.
Curr Biol ; 29(24): 4155-4168.e5, 2019 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-31761703

RESUMEN

Among the neuronal populations implicated in sleep-wake control, the ventrolateral preoptic (VLPO) nucleus has emerged as a key sleep-promoting center. However, the synaptic drives that regulate the VLPO to control arousal levels in vivo have not to date been identified. Here, we show that sleep-promoting galaninergic neurons within the VLPO nucleus, defined pharmacologically and by single-cell transcript analysis, are postsynaptic targets of lateral hypothalamic GABAergic (LHGABA) neurons and that activation of this pathway in vivo rapidly drives wakefulness. Ca2+ imaging from LHGABA neurons indicate that they are both wake and rapid eye movement (REM)-sleep active. Consistent with the potent arousal-promoting property of the LHGABA → VLPO pathway, presynaptic inputs to LHGABA neurons originate from several canonical stress- and arousal-related network nodes. This work represents the first demonstration that direct synaptic inhibition of the VLPO area can suppress sleep-promoting neurons to rapidly promote arousal.


Asunto(s)
Área Preóptica/metabolismo , Sueño/fisiología , Vigilia/fisiología , Animales , Nivel de Alerta/fisiología , Encéfalo/fisiología , Electroencefalografía/métodos , Femenino , Neuronas GABAérgicas/metabolismo , Área Hipotalámica Lateral/fisiología , Hipotálamo/fisiología , Masculino , Ratones , Neuronas/fisiología , Área Preóptica/fisiología , Trastornos del Sueño del Ritmo Circadiano/fisiopatología
14.
J Neurosci ; 39(45): 8929-8939, 2019 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-31548232

RESUMEN

The histaminergic neurons of the tuberomammillary nucleus (TMNHDC) of the posterior hypothalamus have long been implicated in promoting arousal. More recently, a role for GABAergic signaling by the TMNHDC neurons in arousal control has been proposed. Here, we investigated the effects of selective chronic disruption of GABA synthesis (via genetic deletion of the GABA synthesis enzyme, glutamic acid decarboxylase 67) or GABAergic transmission (via genetic deletion of the vesicular GABA transporter (VGAT)) in the TMNHDC neurons on sleep-wake in male mice. We also examined the effects of acute chemogenetic activation and optogenetic inhibition of TMNHDC neurons upon arousal in male mice. Unexpectedly, we found that neither disruption of GABA synthesis nor GABAergic transmission altered hourly sleep-wake quantities, perhaps because very few TMNHDC neurons coexpressed VGAT. Acute chemogenetic activation of TMNHDC neurons did not increase arousal levels above baseline but did enhance vigilance when the mice were exposed to a behavioral cage change challenge. Similarly, acute optogenetic inhibition had little effect upon baseline levels of arousal. In conclusion, we could not identify a role for GABA release by TMNHDC neurons in arousal control. Further, if TMNHDC neurons do release GABA, the mechanism by which they do so remains unclear. Our findings support the view that TMNHDC neurons may be important for enhancing arousal under certain conditions, such as exposure to a novel environment, but play only a minor role in behavioral and EEG arousal under baseline conditions.SIGNIFICANCE STATEMENT The histaminergic neurons of the tuberomammillary nucleus of the hypothalamus (TMNHDC) have long been thought to promote arousal. Additionally, TMNHDC neurons may counter-regulate the wake-promoting effects of histamine through co-release of the inhibitory neurotransmitter, GABA. Here, we show that impairing GABA signaling from TMNHDC neurons does not impact sleep-wake amounts and that few TMNHDC neurons contain the vesicular GABA transporter, which is presumably required to release GABA. We further show that acute activation or inhibition of TMNHDC neurons has limited effects upon baseline arousal levels and that activation enhances vigilance during a behavioral challenge. Counter to general belief, our findings support the view that TMNHDC neurons are neither necessary nor sufficient for the initiation and maintenance of arousal under baseline conditions.


Asunto(s)
Nivel de Alerta , Histamina/metabolismo , Área Hipotalámica Lateral/fisiología , Neuronas/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Potenciales de Acción , Animales , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/metabolismo , Área Hipotalámica Lateral/citología , Área Hipotalámica Lateral/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/fisiología , Sueño , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/genética , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo
15.
Science ; 363(6429): 880-884, 2019 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-30679382

RESUMEN

The sleep-wake cycle regulates interstitial fluid (ISF) and cerebrospinal fluid (CSF) levels of ß-amyloid (Aß) that accumulates in Alzheimer's disease (AD). Furthermore, chronic sleep deprivation (SD) increases Aß plaques. However, tau, not Aß, accumulation appears to drive AD neurodegeneration. We tested whether ISF/CSF tau and tau seeding and spreading were influenced by the sleep-wake cycle and SD. Mouse ISF tau was increased ~90% during normal wakefulness versus sleep and ~100% during SD. Human CSF tau also increased more than 50% during SD. In a tau seeding-and-spreading model, chronic SD increased tau pathology spreading. Chemogenetically driven wakefulness in mice also significantly increased both ISF Aß and tau. Thus, the sleep-wake cycle regulates ISF tau, and SD increases ISF and CSF tau as well as tau pathology spreading.


Asunto(s)
Encéfalo/metabolismo , Ritmo Circadiano , Líquido Extracelular/química , Privación de Sueño/metabolismo , Sueño/fisiología , Vigilia/fisiología , Proteínas tau/análisis , Proteínas tau/líquido cefalorraquídeo , Péptidos beta-Amiloides/análisis , Péptidos beta-Amiloides/líquido cefalorraquídeo , Péptidos beta-Amiloides/metabolismo , Animales , Líquido Extracelular/metabolismo , Femenino , Masculino , Ratones , Ratones Transgénicos , Privación de Sueño/líquido cefalorraquídeo , Vigilia/genética , Proteínas tau/metabolismo
16.
Neuropharmacology ; 154: 34-49, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30503993

RESUMEN

The lateral hypothalamus (LH) is a functionally and anatomically complex brain region that is involved in the regulation of many behavioral and physiological processes including feeding, arousal, energy balance, stress, reward and motivated behaviors, pain perception, body temperature regulation, digestive functions and blood pressure. Despite noteworthy experimental efforts over the past decades, the circuit, cellular and synaptic bases by which these different processes are regulated by the LH remains incompletely understood. This knowledge gap links in large part to the high cellular heterogeneity of the LH. Fortunately, the rapid evolution of newer genetic and electrophysiological tools is now permitting the selective manipulation, typically genetically-driven, of discrete LH cell populations. This, in turn, permits not only assignment of function to discrete cell groups, but also reveals that considerable synergistic and antagonistic interactions exist between key LH cell populations that regulate feeding and arousal. For example, we now know that while LH melanin-concentrating hormone (MCH) and orexin/hypocretin neurons both function as sensors of the internal metabolic environment, their roles regulating sleep and arousal are actually opposing. Additional studies have uncovered similarly important roles for subpopulations of LH GABAergic cells in the regulation of both feeding and arousal. Herein we review the role of LH MCH, orexin/hypocretin and GABAergic cell populations in the regulation of energy homeostasis (including feeding) and sleep-wake and discuss how these three cell populations, and their subpopulations, may interact to optimize and coordinate metabolism, sleep and arousal. This article is part of the Special Issue entitled 'Hypothalamic Control of Homeostasis'.


Asunto(s)
Ingestión de Alimentos/fisiología , Metabolismo Energético/fisiología , Área Hipotalámica Lateral/metabolismo , Sueño/fisiología , Vigilia/fisiología , Animales , Humanos , Vías Nerviosas/metabolismo , Orexinas/metabolismo
17.
Endocrinology ; 159(9): 3158-3176, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30010830

RESUMEN

The lateral hypothalamic area (LHA) is essential for motivated ingestive and locomotor behaviors that impact body weight, yet it remains unclear how the neurochemically defined subpopulations of LHA neurons contribute to energy balance. In particular, the role of the large population of LHA neurotensin (Nts) neurons has remained ambiguous due to the lack of methods to easily visualize and modulate these neurons. Because LHA Nts neurons are activated by leptin and other anorectic cues and they modulate dopamine or local LHA orexin neurons implicated in energy balance, they may have important, unappreciated roles for coordinating behaviors necessary for proper body weight. In this study, we genetically ablated or chemogenetically inhibited LHA Nts neurons in adult mice to determine their necessity for control of motivated behaviors and body weight. Genetic ablation of LHA Nts neurons resulted in profoundly increased adiposity compared with mice with intact LHA Nts neurons, as well as diminished locomotor activity, energy expenditure, and water intake. Complete loss of LHA Nts neurons also led to downregulation of orexin, revealing important cross-talk between the LHA Nts and orexin populations in maintenance of behavior and body weight. In contrast, chemogenetic inhibition of intact LHA Nts neurons did not disrupt orexin expression, but it suppressed locomotor activity and the adaptive response to leptin. Taken together, these data reveal the necessity of LHA Nts neurons and their activation for controlling energy balance, and that LHA Nts neurons influence behavior and body weight via orexin-dependent and orexin-independent mechanisms.


Asunto(s)
Adiposidad/fisiología , Peso Corporal/fisiología , Metabolismo Energético/fisiología , Conducta Alimentaria/fisiología , Área Hipotalámica Lateral/citología , Locomoción/fisiología , Neuronas/fisiología , Neurotensina/metabolismo , Orexinas/metabolismo , Adaptación Fisiológica , Animales , Conducta Animal/fisiología , Regulación hacia Abajo , Conducta de Ingestión de Líquido/fisiología , Expresión Génica , Leptina/metabolismo , Ratones , Neuronas/metabolismo , Obesidad/metabolismo
18.
Curr Biol ; 28(14): 2291-2301.e5, 2018 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-30017482

RESUMEN

Stress elicits a variety of autonomic responses, including hyperthermia (stress fever) in humans and animals. In this present study, we investigated the circuit basis for thermogenesis and heat conservation during this response. We first demonstrated the glutamatergic identity of the dorsal hypothalamic area (DHAVglut2) neurons that innervate the raphe pallidus nucleus (RPa) to regulate core temperature (Tc) and mediate stress-induced hyperthermia. Then, using chemogenetic and optogenetic methods to manipulate this hypothalamomedullary circuit, we found that activation of DHAVglut2 neurons potently drove an increase in Tc, but surprisingly, stress-induced hyperthermia was only reduced by about one-third when they were inhibited. Further investigation showed that DHAVglut2 neurons activate brown adipose tissue (BAT) but do not cause vasoconstriction, instead allowing reflex tail artery vasodilation as a response to BAT-induced hyperthermia. Retrograde rabies virus tracing revealed projections from DHAVglut2 neurons to RPaVglut3, but not to RPaGABA neurons, and identified a set of inputs to DHAVglut2 → RPa neurons that are likely to mediate BAT activation. The dissociation of the DHAVglut2 thermogenic pathway from the thermoregulatory vasoconstriction (heat-conserving) pathway may explain stress flushing (skin vasodilation but a feeling of being too hot) during stressful times.


Asunto(s)
Regulación de la Temperatura Corporal/fisiología , Fiebre/fisiopatología , Hipotálamo/metabolismo , Neuronas/fisiología , Termogénesis , Animales , Femenino , Masculino , Ratones , Núcleo Pálido del Rafe/fisiología , Optogenética , Estrés Fisiológico
19.
J Neurosci ; 38(22): 5168-5181, 2018 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-29735555

RESUMEN

Recent studies have identified an especially important role for basal forebrain GABAergic (BFVGAT) neurons in the regulation of behavioral waking and fast cortical rhythms associated with cognition. However, BFVGAT neurons comprise several neurochemically and anatomically distinct subpopulations, including parvalbumin-containing BFVGAT neurons and somatostatin-containing BFVGAT neurons (BFSOM neurons), and it was recently reported that optogenetic activation of BFSOM neurons increases the probability of a wakefulness to non-rapid-eye movement (NREM) sleep transition when stimulated during the rest period of the animal. This finding was unexpected given that most BFSOM neurons are not NREM sleep active and that central administration of the synthetic somatostatin analog, octreotide, suppresses NREM sleep or increases REM sleep. Here we used a combination of genetically driven chemogenetic and optogenetic activation, chemogenetic inhibition, and ablation approaches to further explore the in vivo role of BFSOM neurons in arousal control. Our findings indicate that acute activation or inhibition of BFSOM neurons is neither wakefulness nor NREM sleep promoting and is without significant effect on the EEG, and that chronic loss of these neurons is without effect on total 24 h sleep amounts, although a small but significant increase in waking was observed in the lesioned mice during the early active period. Our in vitro cell recordings further reveal electrophysiological heterogeneity in BFSOM neurons, specifically suggesting at least two distinct subpopulations. Together, our data support the more nuanced view that BFSOM neurons are electrically heterogeneous and are not NREM sleep or wake promoting per se, but may exert, in particular during the early active period, a modest inhibitory influence on arousal circuitry.SIGNIFICANCE STATEMENT The cellular basal forebrain (BF) is a highly complex area of the brain that is implicated in a wide range of higher-level neurobiological processes, including regulating and maintaining normal levels of electrocortical and behavioral arousal. The respective in vivo roles of BF cell populations and their neurotransmitter systems in the regulation of electrocortical and behavioral arousal remains incompletely understood. Here we seek to define the neurobiological contribution of GABAergic somatostatin-containing BF neurons to arousal control. Understanding the respective contribution of BF cell populations to arousal control may provide critical insight into the pathogenesis of a host of neuropsychiatric and neurodegenerative disorders, including Alzheimer's disease, Parkinson's disease, schizophrenia, and the cognitive impairments of normal aging.


Asunto(s)
Prosencéfalo Basal/fisiología , Conducta Animal/fisiología , Neuronas/fisiología , Somatostatina/fisiología , Animales , Prosencéfalo Basal/citología , Electroencefalografía , Fenómenos Electrofisiológicos/fisiología , Femenino , Eliminación de Gen , Genotipo , Masculino , Ratones , Optogenética , Sueño de Onda Lenta/fisiología , Somatostatina/metabolismo , Activación Transcripcional , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/genética , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/fisiología , Vigilia/fisiología
20.
Nat Neurosci ; 21(5): 717-724, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29632359

RESUMEN

'Sundowning' in dementia and Alzheimer's disease is characterized by early-evening agitation and aggression. While such periodicity suggests a circadian origin, whether the circadian clock directly regulates aggressive behavior is unknown. We demonstrate that a daily rhythm in aggression propensity in male mice is gated by GABAergic subparaventricular zone (SPZGABA) neurons, the major postsynaptic targets of the central circadian clock, the suprachiasmatic nucleus. Optogenetic mapping revealed that SPZGABA neurons receive input from vasoactive intestinal polypeptide suprachiasmatic nucleus neurons and innervate neurons in the ventrolateral part of the ventromedial hypothalamus (VMH), which is known to regulate aggression. Additionally, VMH-projecting dorsal SPZ neurons are more active during early day than early night, and acute chemogenetic inhibition of SPZGABA transmission phase-dependently increases aggression. Finally, SPZGABA-recipient central VMH neurons directly innervate ventrolateral VMH neurons, and activation of this intra-VMH circuit drove attack behavior. Altogether, we reveal a functional polysynaptic circuit by which the suprachiasmatic nucleus clock regulates aggression.


Asunto(s)
Agresión/fisiología , Ritmo Circadiano/fisiología , Hipotálamo/fisiología , Vías Nerviosas/fisiología , Animales , Mapeo Encefálico , Corticosterona/sangre , Potenciales Postsinápticos Excitadores/fisiología , Hipotálamo/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Vías Nerviosas/citología , Optogenética , Núcleo Hipotalámico Paraventricular/citología , Núcleo Hipotalámico Paraventricular/fisiología , Núcleo Supraquiasmático/fisiología , Péptido Intestinal Vasoactivo/fisiología , Núcleo Hipotalámico Ventromedial/citología , Núcleo Hipotalámico Ventromedial/fisiología , Ácido gamma-Aminobutírico/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...