Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 4475, 2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38796568

RESUMEN

About half of the neurons in the parabrachial nucleus (PB) that are activated by CO2 are located in the external lateral (el) subnucleus, express calcitonin gene-related peptide (CGRP), and cause forebrain arousal. We report here, in male mice, that most of the remaining CO2-responsive neurons in the adjacent central lateral (PBcl) and Kölliker-Fuse (KF) PB subnuclei express the transcription factor FoxP2 and many of these neurons project to respiratory sites in the medulla. PBclFoxP2 neurons show increased intracellular calcium during wakefulness and REM sleep and in response to elevated CO2 during NREM sleep. Photo-activation of the PBclFoxP2 neurons increases respiration, whereas either photo-inhibition of PBclFoxP2 or genetic deletion of PB/KFFoxP2 neurons reduces the respiratory response to CO2 stimulation without preventing awakening. Thus, augmenting the PBcl/KFFoxP2 response to CO2 in patients with sleep apnea in combination with inhibition of the PBelCGRP neurons may avoid hypoventilation and minimize EEG arousals.


Asunto(s)
Dióxido de Carbono , Factores de Transcripción Forkhead , Hipercapnia , Neuronas , Núcleos Parabraquiales , Vigilia , Animales , Hipercapnia/fisiopatología , Hipercapnia/metabolismo , Neuronas/metabolismo , Neuronas/fisiología , Masculino , Núcleos Parabraquiales/fisiología , Núcleos Parabraquiales/metabolismo , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/genética , Ratones , Dióxido de Carbono/metabolismo , Vigilia/fisiología , Respiración , Ratones Endogámicos C57BL , Péptido Relacionado con Gen de Calcitonina/metabolismo , Sueño REM/fisiología , Proteínas Represoras
2.
Res Sq ; 2023 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-37205518

RESUMEN

Many species use a temporary drop in body temperature and metabolic rate (torpor) as a strategy to survive food scarcity. A similar profound hypothermia is observed with activation of preoptic neurons that express the neuropeptides Pituitary Adenylate-Cyclase-Activating Polypeptide (PACAP)1, Brain Derived Neurotrophic Factor (BDNF)2, or Pyroglutamylated RFamide Peptide (QRFP)3, the vesicular glutamate transporter, Vglut24,5 or the leptin receptor6 (LepR), estrogen 1 receptor (Esr1)7 or prostaglandin E receptor 3 (EP3R) in mice8. However, most of these genetic markers are found on multiple populations of preoptic neurons and only partially overlap with one another. We report here that expression of the EP3R marks a unique population of median preoptic (MnPO) neurons that are required both for lipopolysaccharide (LPS)-induced fever9 and for torpor. These MnPOEP3R neurons produce persistent fever responses when inhibited and prolonged hypothermic responses when activated either chemo- or opto-genetically even for brief periods of time. The mechanism for these prolonged responses appears to involve increases in intracellular calcium in individual EP3R-expressing preoptic neurons that persist for many minutes up to hours beyond the termination of a brief stimulus. These properties endow MnPOEP3R neurons with the ability to act as a two-way master switch for thermoregulation.

3.
Sleep ; 45(12)2022 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-36170177

RESUMEN

STUDY OBJECTIVES: The pedunculopontine tegmental (PPT) nucleus is implicated in many brain functions, ranging from sleep/wake control and locomotion, to reward mechanisms and learning. The PPT contains cholinergic, GABAergic, and glutamatergic neurons with extensive ascending and descending axonal projections. Glutamatergic PPT (PPTvGlut2) neurons are thought to promote wakefulness, but the mechanisms through which this occurs are unknown. In addition, some researchers propose that PPTvGlut2 neurons promote locomotion, yet even though the PPT is a target for deep brain stimulation in Parkinson's disease, the role of the PPT in locomotion is debated. We hypothesized that PPTvGluT2 neurons drive arousal and specific waking behaviors via certain projections and modulate locomotion via others. METHODS: We mapped the axonal projections of PPTvGlut2 neurons using conditional anterograde tracing and then photostimulated PPTvGlut2 soma or their axon terminal fields across sleep/wake states and analyzed sleep/wake behavior, muscle activity, and locomotion in transgenic mice. RESULTS: We found that stimulation of PPTvGlut2 soma and their axon terminals rapidly triggered arousals from non-rapid eye movement sleep, especially with activation of terminals in the basal forebrain (BF) and lateral hypothalamus (LH). With photoactivation of PPTvGlut2 terminals in the BF and LH, this wakefulness was accompanied by locomotion and other active behaviors, but stimulation of PPTvGlut2 soma and terminals in the substantia nigra triggered only quiet wakefulness without locomotion. CONCLUSIONS: These findings demonstrate the importance of the PPTvGluT2 neurons in driving various aspects of arousal and show that heterogeneous brain nuclei, such as the PPT, can promote a variety of behaviors via distinct axonal projections.


Asunto(s)
Prosencéfalo Basal , Vigilia , Animales , Ratones , Vigilia/fisiología , Neuronas/fisiología , Sueño/fisiología , Axones
4.
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
5.
Metabolism ; 130: 155158, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35150732

RESUMEN

BACKGROUND: Nearly 14% of Americans experience chronic circadian disruption due to shift work, increasing their risk of obesity, diabetes, and other cardiometabolic disorders. These disorders are also exacerbated by modern eating habits such as frequent snacking and consumption of high-fat foods. METHODS: We investigated the effects of recurrent circadian disruption (RCD) on glucose metabolism in C57BL/6 mice and in human participants exposed to non-24-h light-dark (LD) schedules vs. those on standard 24-h LD schedules. These LD schedules were designed to induce circadian misalignment between behaviors including rest/activity and fasting/eating with the output of the near-24-h central circadian pacemaker, while minimizing sleep loss, and were maintained for 12 weeks in mice and 3 weeks in humans. We examined interactions of these circadian-disrupted schedules compared to control 24-h schedules with a lower-fat diet (LFD, 13% in mouse and 25-27% in humans) and high-fat diet (HFD, 45% in mouse and 45-50% in humans). We also used young vs. older mice to determine whether they would respond differently to RCD. RESULTS: When combined with a HFD, we found that RCD caused significant weight gain in mice and increased body fat in humans, and significantly impaired glucose tolerance and insulin sensitivity in both mice and humans, but this did not occur when RCD was combined with a LFD. This effect was similar in both young and older mice. CONCLUSION: These results in both humans and a model organism indicate that circadian disruption has an adverse effect on metabolism among individuals eating a high-fat Western-style diet, even in the absence of significant sleep loss, and suggest that reducing dietary fat may protect against the metabolic consequences of a lifestyle (such as shift work) that involves chronic circadian disruption.


Asunto(s)
Dieta Alta en Grasa , Insulina , Animales , Dieta Alta en Grasa/efectos adversos , Glucosa , Humanos , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología
6.
Curr Neurol Neurosci Rep ; 20(12): 55, 2020 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-33006677

RESUMEN

PURPOSE OF THE REVIEW: Melanin-concentrating hormone (MCH)-expressing neurons located in the lateral hypothalamus are considered as an integral component of sleep-wake circuitry. However, the precise role of MCH neurons in sleep-wake regulation has remained unclear, despite several years of research employing a wide range of techniques. We review recent data on this aspect, which are mostly inconsistent, and propose a novel role for MCH neurons in sleep regulation. RECENT FINDINGS: While almost all studies using "gain-of-function" approaches show an increase in rapid eye movement sleep (or paradoxical sleep; PS), loss-of-function approaches have not shown reductions in PS. Similarly, the reported changes in wakefulness or non-rapid eye movement sleep (slow-wave sleep; SWS) with manipulation of the MCH system using conditional genetic methods are inconsistent. Currently available data do not support a role for MCH neurons in spontaneous sleep-wake but imply a crucial role for them in orchestrating sleep-wake responses to changes in external and internal environments.


Asunto(s)
Hormonas Hipotalámicas , Humanos , Hormonas Hipotalámicas/genética , Melaninas , Neuronas , Hormonas Hipofisarias/genética , Sueño , Vigilia
7.
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
8.
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
9.
J Neurosci ; 40(12): 2573-2588, 2020 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-32079648

RESUMEN

Fever is a common phenomenon during infection or inflammatory conditions. This stereotypic rise in body temperature (Tb) in response to inflammatory stimuli is a result of autonomic responses triggered by prostaglandin E2 action on EP3 receptors expressed by neurons in the median preoptic nucleus (MnPOEP3R neurons). To investigate the identity of MnPOEP3R neurons, we first used in situ hybridization to show coexpression of EP3R and the VGluT2 transporter in MnPO neurons. Retrograde tracing showed extensive direct projections from MnPOVGluT2 but few from MnPOVgat neurons to a key site for fever production, the raphe pallidus. Ablation of MnPOVGluT2 but not MnPOVgat neurons abolished fever responses but not changes in Tb induced by behavioral stress or thermal challenges. Finally, we crossed EP3R conditional knock-out mice with either VGluT2-IRES-cre or Vgat-IRES-cre mice and used both male and female mice to confirm that the neurons that express EP3R and mediate fever are glutamatergic, not GABAergic. This finding will require rethinking current concepts concerning the central thermoregulatory pathways based on the MnPOEP3R neurons being GABAergic.SIGNIFICANCE STATEMENT Body temperature is regulated by the CNS. The rise of the body temperature, or fever, is an important brain-orchestrated mechanism for fighting against infectious or inflammatory disease, and is tightly regulated by the neurons located in the median preoptic nucleus (MnPO). Here we demonstrate that excitatory MnPO neurons mediate fever and examine a potential central circuit underlying the development of fever responses.


Asunto(s)
Fiebre/fisiopatología , Ácido Glutámico , Inflamación/fisiopatología , Neuronas , Área Preóptica/fisiopatología , Subtipo EP3 de Receptores de Prostaglandina E , Animales , Temperatura Corporal , Regulación de la Temperatura Corporal , Femenino , Fiebre/inducido químicamente , Globo Pálido/fisiopatología , Inflamación/inducido químicamente , Lipopolisacáridos , Masculino , Ratones , Ratones Noqueados , Actividad Motora , Vías Nerviosas/fisiopatología , Área Preóptica/citología , Estrés Psicológico , Proteína 2 de Transporte Vesicular de Glutamato/genética
10.
J Comp Neurol ; 528(3): 380-388, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31454077

RESUMEN

Accumulating evidence supports the role of sleep in synaptic plasticity and memory consolidation. One line of investigation, the synaptic homeostasis hypothesis, has emphasized the increase in synaptic strength during waking, and compensatory downsizing of (presumably less frequently used) synapses during sleep. Conversely, other studies have reported downsizing and loss of dendritic spines following sleep deprivation. We wanted to determine the effect of sleep deprivation on dendritic spines of hippocampal CA1 neurons using genetic methods for fluorescent labeling of dendritic spines. Male Vglut2-Cre mice were injected with an AAV-DIO-ChR2-mCherry reporter in CA1 hippocampus. Gentle handling was used to sleep deprive mice for 5 hr, from lights on (7 am) to 12 noon. Control and sleep-deprived mice were euthanized at 12 noon and processed for quantification of dendritic spines. We used confocal microscope imaging and three-dimensional (3D) analysis to quantify thin, mushroom, and stubby spines from CA1 dendrites, distinguishing between branch segments. We observed significantly greater density of spines in CA1 of sleep-deprived mice, driven primarily by greater numbers of thin spines, and significantly larger spine volume and head diameter. Branch and region-specific analysis revealed that spine volume was greater in primary dendrites of apical and basal segments, along with proximal segments on both apical and basal dendrites, and spine density was increased in secondary branches and distal segments on apical dendrites following sleep deprivation. Our 3D quantification suggests sleep contributes to region- and branch-specific synaptic downscaling in the hippocampus, supporting the theory of broad but selective synaptic downscaling during sleep.


Asunto(s)
Espinas Dendríticas/fisiología , Hipocampo/fisiopatología , Privación de Sueño/fisiopatología , Animales , Espinas Dendríticas/patología , Hipocampo/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Privación de Sueño/patología
11.
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
12.
PLoS Biol ; 17(3): e3000172, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30893297

RESUMEN

Sleep and wakefulness are greatly influenced by various physiological and psychological factors, but the neuronal elements responsible for organizing sleep-wake behavior in response to these factors are largely unknown. In this study, we report that a subset of neurons in the lateral hypothalamic area (LH) expressing the neuropeptide neurotensin (Nts) is critical for orchestrating sleep-wake responses to acute psychological and physiological challenges or stressors. We show that selective activation of NtsLH neurons with chemogenetic or optogenetic methods elicits rapid transitions from non-rapid eye movement (NREM) sleep to wakefulness and produces sustained arousal, higher locomotor activity (LMA), and hyperthermia, which are commonly observed after acute stress exposure. On the other hand, selective chemogenetic inhibition of NtsLH neurons attenuates the arousal, LMA, and body temperature (Tb) responses to a psychological stress (a novel environment) and augments the responses to a physiological stress (fasting).


Asunto(s)
Fiebre/metabolismo , Área Hipotalámica Lateral/metabolismo , Neurotensina/metabolismo , Animales , Temperatura Corporal , Electroforesis , Técnicas de Genotipaje , Locomoción/fisiología , Masculino , Ratones , Neuronas/metabolismo
13.
Neuroscience ; 406: 314-324, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30890480

RESUMEN

Neurons containing melanin-concentrating hormone (MCH) in the lateral hypothalamic area (LH) have been shown to promote rapid eye movement sleep (REMs) in mice. However, the downstream neural pathways through which MCH neurons influence REMs remained unclear. Because MCH neurons are considered to be primarily inhibitory, we hypothesized that these neurons inhibit the midbrain 'REMs-suppressing' region consisting of the ventrolateral periaqueductal gray and the lateral pontine tegmentum (vlPAG/LPT) to promote REMs. To test this hypothesis, we optogenetically inhibited MCH terminals in the vlPAG/LPT under baseline conditions as well as with simultaneous chemogenetic activation of MCH soma. We found that inhibition of MCH terminals in the vlPAG/LPT significantly reduced transitions into REMs during spontaneous sleep-wake cycles and prevented the increase in REMs transitions observed after chemogenetic activation of MCH neurons. These results strongly suggest that the vlPAG/LPT may be an essential relay through which MCH neurons modulate REMs.


Asunto(s)
Movimientos Oculares/fisiología , Hormonas Hipotalámicas/metabolismo , Melaninas/metabolismo , Sustancia Gris Periacueductal/fisiología , Hormonas Hipofisarias/metabolismo , Sueño REM/fisiología , Animales , Área Hipotalámica Lateral/fisiología , Masculino , Ratones Transgénicos , Vías Nerviosas/fisiología , Neuronas/fisiología , Vigilia/fisiología
14.
Brain Struct Funct ; 224(1): 99-110, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30284033

RESUMEN

Neurons containing melanin-concentrating hormone (MCH) in the posterior lateral hypothalamus play an integral role in rapid eye movement sleep (REMs) regulation. As MCH neurons also contain a variety of other neuropeptides [e.g., cocaine- and amphetamine-regulated transcript (CART) and nesfatin-1] and neurotransmitters (e.g., glutamate), the specific neurotransmitter responsible for REMs regulation is not known. We hypothesized that glutamate, the primary fast-acting neurotransmitter in MCH neurons, is necessary for REMs regulation. To test this hypothesis, we deleted vesicular glutamate transporter (Vglut2; necessary for synaptic release of glutamate) specifically from MCH neurons by crossing MCH-Cre mice (expressing Cre recombinase in MCH neurons) with Vglut2flox/flox mice (expressing LoxP-modified alleles of Vglut2), and studied the amounts, architecture and diurnal variation of sleep-wake states during baseline conditions. We then activated the MCH neurons lacking glutamate neurotransmission using chemogenetic methods and tested whether these MCH neurons still promoted REMs. Our results indicate that glutamate in MCH neurons contributes to normal diurnal variability of REMs by regulating the levels of REMs during the dark period, but MCH neurons can promote REMs even in the absence of glutamate.


Asunto(s)
Ritmo Circadiano , Ácido Glutámico/metabolismo , Hormonas Hipotalámicas/metabolismo , Hipotálamo Posterior/metabolismo , Melaninas/metabolismo , Neuronas/metabolismo , Hormonas Hipofisarias/metabolismo , Sueño REM , Animales , Hormonas Hipotalámicas/genética , Hipotálamo Posterior/citología , Masculino , Melaninas/genética , Ratones Transgénicos , Fotoperiodo , Hormonas Hipofisarias/genética , Factores de Tiempo , Proteína 2 de Transporte Vesicular de Glutamato/genética , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo , Vigilia
15.
Nat Commun ; 9(1): 4129, 2018 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-30297727

RESUMEN

The preoptic area (POA) is necessary for sleep, but the fundamental POA circuits have remained elusive. Previous studies showed that galanin (GAL)- and GABA-producing neurons in the ventrolateral preoptic nucleus (VLPO) express cFos after periods of increased sleep and innervate key wake-promoting regions. Although lesions in this region can produce insomnia, high frequency photostimulation of the POAGAL neurons was shown to paradoxically cause waking, not sleep. Here we report that photostimulation of VLPOGAL neurons in mice promotes sleep with low frequency stimulation (1-4 Hz), but causes conduction block and waking at frequencies above 8 Hz. Further, optogenetic inhibition reduces sleep. Chemogenetic activation of VLPOGAL neurons confirms the increase in sleep, and also reduces body temperature. In addition, chemogenetic activation of VLPOGAL neurons induces short-latency sleep in an animal model of insomnia. Collectively, these findings establish a causal role of VLPOGAL neurons in both sleep induction and heat loss.


Asunto(s)
Regulación de la Temperatura Corporal/fisiología , Galanina/metabolismo , Neuronas/metabolismo , Área Preóptica/metabolismo , Sueño/fisiología , Animales , Regulación de la Temperatura Corporal/genética , Electroencefalografía , Electromiografía , Galanina/genética , Perfilación de la Expresión Génica , Masculino , Ratones Transgénicos , Área Preóptica/citología , Sueño/genética , Trastornos del Inicio y del Mantenimiento del Sueño/genética , Trastornos del Inicio y del Mantenimiento del Sueño/metabolismo , Trastornos del Inicio y del Mantenimiento del Sueño/fisiopatología
16.
Neurobiol Dis ; 120: 12-20, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30149182

RESUMEN

The lateral hypothalamus contains neurons producing orexins that promote wakefulness and suppress REM sleep as well as neurons producing melanin-concentrating hormone (MCH) that likely promote REM sleep. Narcolepsy with cataplexy is caused by selective loss of the orexin neurons, and the MCH neurons appear unaffected. As the orexin and MCH systems exert opposing effects on REM sleep, we hypothesized that imbalance in this REM sleep-regulating system due to activity in the MCH neurons may contribute to the striking REM sleep dysfunction characteristic of narcolepsy. To test this hypothesis, we chemogenetically activated the MCH neurons and pharmacologically blocked MCH signaling in a murine model of narcolepsy and studied the effects on sleep-wake behavior and cataplexy. To chemoactivate MCH neurons, we injected an adeno-associated viral vector containing the hM3Dq stimulatory DREADD into the lateral hypothalamus of orexin null mice that also express Cre recombinase in the MCH neurons (MCH-Cre::OX-KO mice) and into control MCH-Cre mice with normal orexin expression. In both lines of mice, activation of MCH neurons by clozapine-N-oxide (CNO) increased rapid eye movement (REM) sleep without altering other states. In mice lacking orexins, activation of the MCH neurons also increased abnormal intrusions of REM sleep manifest as cataplexy and short latency transitions into REM sleep (SLREM). Conversely, a MCH receptor 1 antagonist, SNAP 94847, almost completely eliminated SLREM and cataplexy in OX-KO mice. These findings affirm that MCH neurons promote REM sleep under normal circumstances, and their activity in mice lacking orexins likely triggers abnormal intrusions of REM sleep into non-REM sleep and wake, resulting in the SLREM and cataplexy characteristic of narcolepsy.


Asunto(s)
Hormonas Hipotalámicas/metabolismo , Melaninas/metabolismo , Narcolepsia/metabolismo , Neuronas/fisiología , Hormonas Hipofisarias/metabolismo , Sueño REM/fisiología , Animales , Femenino , Hormonas Hipotalámicas/antagonistas & inhibidores , Hormonas Hipotalámicas/genética , Masculino , Melaninas/antagonistas & inhibidores , Melaninas/genética , Ratones , Ratones Noqueados , Narcolepsia/genética , Neuronas/efectos de los fármacos , Piperidinas/farmacología , Hormonas Hipofisarias/antagonistas & inhibidores , Hormonas Hipofisarias/genética
17.
Case Rep Hematol ; 2017: 1850821, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29158925

RESUMEN

Cytomegalovirus (CMV) is a type of herpes infection that has a characteristic feature of maintaining lifelong latency within the host cell. CMV manifestations can cover a broad spectrum from fever to as severe as pancytopenia, hepatitis, retinitis, meningoencephalitis, Guillain-Barre syndrome, pneumonia, and thrombosis. Multiple case reports of thrombosis associated with CMV have been reported. Deep vein thrombosis or pulmonary embolism is more common in immunocompetent patients while splenic infarct is more common in immunocompromised patients. However, here we report a female patient on low-dose methotrexate for rheumatoid arthritis who presented with both pulmonary embolism and splenic infarct.

18.
Artículo en Inglés | MEDLINE | ID: mdl-28924481

RESUMEN

Euglycemic diabetic ketoacidosis (EDKA) is a clinical triad comprising increased anion gap metabolic acidosis, ketonemia or ketonuria and normal blood glucose levels <200 mg/dL. This condition is a diagnostic challenge as euglycemia masquerades the underlying diabetic ketoacidosis. Thus, a high clinical suspicion is warranted, and other diagnosis ruled out. Here, we present two patients on regular insulin treatment who were admitted with a diagnosis of EDKA. The first patient had insulin pump failure and the second patient had urinary tract infection and nausea, thereby resulting in starvation. Both of them were aggressively treated with intravenous fluids and insulin drip as per the protocol for the blood glucose levels till the anion gap normalized, and the metabolic acidosis reversed. This case series summarizes, in brief, the etiology, pathophysiology and treatment of EDKA. LEARNING POINTS: Euglycemic diabetic ketoacidosis is rare.Consider ketosis in patients with DKA even if their serum glucose levels are normal.High clinical suspicion is required to diagnose EDKA as normal blood sugar levels masquerade the underlying DKA and cause a diagnostic and therapeutic dilemma.Blood pH and blood or urine ketones should be checked in ill patients with diabetes regardless of blood glucose levels.

19.
Respir Physiol Neurobiol ; 244: 41-50, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28711601

RESUMEN

Neural mechanisms of obstructive sleep apnea, a common sleep-related breathing disorder, are incompletely understood. Hypoglossal motoneurons, which provide tonic and inspiratory activation of genioglossus (GG) muscle (a major upper airway dilator), receive catecholaminergic input from medullary A1/C1 neurons. We aimed to determine the contribution of A1/C1 neurons in control of GG muscle during sleep and wakefulness. To do so, we placed injections of a viral vector into DBH-cre mice to selectively express the hMD4i inhibitory chemoreceptors in A1/C1 neurons. Administration of the hM4Di ligand, clozapine-N-oxide (CNO), in these mice decreased GG muscle activity during NREM sleep (F1,1,3=17.1, p<0.05); a similar non-significant decrease was observed during wakefulness. CNO administration had no effect on neck muscle activity, respiratory parameters or state durations. In addition, CNO-induced inhibition of A1/C1 neurons did not alter the magnitude of the naturally occurring depression of GG activity during transitions from wakefulness to NREM sleep. These findings suggest that A1/C1 neurons have a net excitatory effect on GG activity that is most likely mediated by hypoglossal motoneurons. However, the activity of A1/C1 neurons does not appear to contribute to NREM sleep-related inhibition of GG muscle activity, suggesting that A1/C1 neurons regulate upper airway patency in a state-independent manner.


Asunto(s)
Catecolaminas/metabolismo , Nervio Hipogloso/fisiología , Bulbo Raquídeo/fisiología , Músculos Respiratorios/fisiología , Fases del Sueño/fisiología , Vigilia/fisiología , Animales , Electroencefalografía , Electromiografía , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas Motoras/fisiología , Músculos del Cuello/inervación , Músculos del Cuello/fisiología , Vías Nerviosas/fisiología , Receptores de Catecolaminas/genética , Receptores de Catecolaminas/metabolismo , Músculos Respiratorios/inervación
20.
Gastroenterology Res ; 10(3): 153-158, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28725301

RESUMEN

While gallstones and alcoholism are widely known to be the most common causative agents of acute pancreatitis, about 10% of cases are thought to be caused by infectious microorganisms. These microorganisms include viruses (e.g. mumps, Coxsackie B, and hepatitis), bacteria (e.g. Mycoplasma pneumoniae and leptospirosis), and parasites (e.g. Ascaris lumbricoides, Fasciola hepatica, and hydatid disease). Each organism causes acute pancreatitis through diverse mechanisms. The review is primarily conducted in an attempt to provide a better understanding of the possibility of acute pancreatitis presenting as a complication relating to these organisms, and the aim is to guide future diagnoses, management, and predictions of complications.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...