Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 13(1): 12299, 2023 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-37516800

RESUMEN

Pharyngeal muscle activity and responsiveness are key pathophysiological traits in human obstructive sleep apnea (OSA) and strong contributors to improvements with pharmacotherapy. The thyrotropin-releasing hormone (TRH) analog taltirelin is of high pre-clinical interest given its neuronal-stimulant properties, minimal endocrine activity, tongue muscle activation following microperfusion into the hypoglossal motor nucleus (HMN) or systemic delivery, and high TRH receptor expression at the HMN compared to rest of the brain. Here we test the hypothesis that taltirelin increases HMN activity and/or responsivity to excitatory stimuli applied across sleep-wake states in-vivo. To target hypoglossal motoneurons with simultaneous pharmacological and optical stimuli we used customized "opto-dialysis" probes and chronically implanted them in mice expressing a light sensitive cation channel exclusively on cholinergic neurons (ChAT-ChR2, n = 12) and wild-type mice lacking the opsin (n = 10). Both optical stimuli applied across a range of powers (P < 0.001) and microperfusion of taltirelin into the HMN (P < 0.020) increased tongue motor activity in sleeping ChAT-ChR2 mice. Notably, taltirelin increased tonic background tongue motor activity (P < 0.001) but not responsivity to excitatory optical stimuli across sleep-wake states (P > 0.098). This differential effect on tonic motor activity versus responsivity informs human studies of the potential beneficial effects of taltirelin on pharyngeal motor control and OSA pharmacotherapy.


Asunto(s)
Diálisis Renal , Apnea Obstructiva del Sueño , Humanos , Animales , Ratones , Neuronas Motoras , Sueño , Cafeína , Colina O-Acetiltransferasa , Niacinamida , Actividad Motora
2.
Expert Opin Ther Targets ; 27(7): 609-626, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37494064

RESUMEN

INTRODUCTION: Obstructive sleep apnea (OSA) is a common and serious breathing disorder. Several pathophysiological factors predispose individuals to OSA. These factors are quantifiable, and modifiable pharmacologically. AREAS COVERED: Four key pharmacotherapeutic targets are identified and mapped to the major determinants of OSA pathophysiology. PubMed and Clinicaltrials.gov were searched through April 2023. EXPERT OPINION: Target #1: Pharyngeal Motor Effectors. Increasing pharyngeal muscle activity and responsivity with noradrenergic-antimuscarinic combination is central to recent breakthrough OSA pharmacotherapy. Assumptions, knowledge gaps, future directions, and other targets are identified. #2: Upper Airway Sensory Afferents. There is translational potential of sensitizing and amplifying reflex pharyngeal dilator muscle responses to negative airway pressure via intranasal delivery of new potassium channel blockers. Rationales, advantages, findings, and potential strategies to enhance effectiveness are identified. #3: Chemosensory Afferents and Ventilatory Control. Strategies to manipulate ventilatory control system sensitivity by carbonic anhydrase inhibitors are supported in theory and initial studies. Intranasal delivery of agents to stimulate central respiratory activity are also introduced. #4: Sleep-Wake Mechanisms. Arousability is the fourth therapeutic target rationalized. Evolving automated tools to measure key pathophysiological factors predisposing to OSA will accelerate pharmacotherapy. Although not currently ready for general clinical settings, the identified targets are of future promise.


Asunto(s)
Apnea Obstructiva del Sueño , Humanos , Apnea Obstructiva del Sueño/tratamiento farmacológico , Sueño/fisiología , Faringe
3.
Elife ; 122023 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-37458576

RESUMEN

Rhythmic breathing is generated by neural circuits located in the brainstem. At its core is the preBötzinger Complex (preBötC), a region of the medulla, necessary for the generation of rhythmic breathing in mammals. The preBötC is comprised of various neuronal populations expressing neurokinin-1 receptors, the cognate G-protein-coupled receptor of the neuropeptide substance P (encoded by the tachykinin precursor 1 or Tac1). Neurokinin-1 receptors are highly expressed in the preBötC and destruction or deletion of neurokinin-1 receptor-expressing preBötC neurons severely impair rhythmic breathing. Although, the application of substance P to the preBötC stimulates breathing in rodents, substance P is also involved in nociception and locomotion in various brain regions, suggesting that Tac1 neurons found in the preBötC may have diverse functional roles. Here, we characterized the role of Tac1-expressing preBötC neurons in the generation of rhythmic breathing in vivo, as well as motor behaviors. Using a cre-lox recombination approach, we injected adeno-associated virus containing the excitatory channelrhodopsin-2 ChETA in the preBötC region of Tac1-cre mice. Employing a combination of histological, optogenetics, respiratory, and behavioral assays, we showed that stimulation of glutamatergic or Tac1 preBötC neurons promoted rhythmic breathing in both anesthetized and freely moving animals, but also triggered locomotion and overcame respiratory depression by opioid drugs. Overall, our study identified a population of excitatory preBötC with major roles in rhythmic breathing and behaviors.


Asunto(s)
Receptores de Neuroquinina-1 , Sustancia P , Ratones , Animales , Receptores de Neuroquinina-1/genética , Neuronas/fisiología , Bulbo Raquídeo/fisiología , Respiración , Centro Respiratorio/fisiología , Mamíferos
4.
Sci Rep ; 12(1): 14896, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-36050440

RESUMEN

Successful cholinergic-noradrenergic pharmacotherapy for obstructive sleep apnea (OSA) is thought to be due to effects at the hypoglossal motor nucleus (HMN). Clinical efficacy varies with muscarinic-receptor (MR) subtype affinities. We hypothesized that oxybutynin (cholinergic agent in successful OSA pharmacotherapy) is an effective MR antagonist at the HMN and characterized its efficacy with other antagonists. We recorded tongue muscle activity of isoflurane anesthetized rats (121 males and 60 females, 7-13 per group across 13 protocols) in response to HMN microperfusion with MR antagonists with and without: (i) eserine-induced increased endogenous acetylcholine at the HMN and (ii) muscarine. Eserine-induced increased acetylcholine decreased tongue motor activity (p < 0.001) with lesser cholinergic suppression in females versus males (p = 0.017). Motor suppression was significantly attenuated by the MR antagonists atropine, oxybutynin, and omadacycline (MR2 antagonist), each p < 0.001, with similar residual activity between agents (p ≥ 0.089) suggesting similar efficacy at the HMN. Sex differences remained with atropine and oxybutynin (p < 0.001 to 0.05) but not omadacycline (p = 0.722). Muscarine at the HMN also decreased motor activity (p < 0.001) but this was not sex-specific (p = 0.849). These findings have translational relevance to antimuscarinic agents in OSA pharmacotherapy and understanding potential sex differences in HMN suppression with increased endogenous acetylcholine related to sparing nicotinic excitation.


Asunto(s)
Nervio Hipogloso , Apnea Obstructiva del Sueño , Acetilcolina/farmacología , Animales , Atropina/farmacología , Femenino , Nervio Hipogloso/fisiología , Masculino , Muscarina/farmacología , Antagonistas Muscarínicos/farmacología , Fisostigmina/farmacología , Ratas , Ratas Wistar
5.
J Appl Physiol (1985) ; 133(5): 1067-1080, 2022 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-36135952

RESUMEN

Taltirelin is a stable, brain-penetrating thyrotropin-releasing hormone (TRH) analog with minimal endocrine activity and potential respiratory stimulant properties. Taltirelin's receptor target shows high differential expression at the hypoglossal motor nucleus, and local taltirelin microperfusion into the hypoglossal motor nucleus causes sustained tongue motor activation compared with the transient activating effects of TRH itself. Here, we performed a randomized, within-subject, repeated-measures design over six separate study days (separated by at least 72 h) in chronically instrumented male (n = 10) and female (n = 9) rats to identify effects on sleep and breathing. Vehicle controls or taltirelin (0.1 and 1 mg/kg) with and without trazodone (30 mg/kg) were administered by intraperitoneal injection. Trazodone was included due to clinical interest in the context of sleep apnea pharmacotherapy as it can suppress arousal without compromising pharyngeal muscle activity. Systemically administered taltirelin (1 but not 0.1 mg/kg) increased tonic and within-breath phasic tonic muscle activity compared with vehicle controls (P ≤ 0.007), with little or no changes in diaphragm amplitude or respiratory rate. Taltirelin also suppressed nonrapid eye movement (non-REM) sleep and increased wakefulness (P ≤ 0.037). Other indices of taltirelin-induced central nervous system arousal included increased trapezius muscle tone in non-REM sleep and decreased total electroencephalogram power and δ (0.5-4 Hz) power (P ≤ 0.046). These effects were especially apparent in non-REM sleep and not prevented by trazodone. These preclinical findings identify taltirelin as a stable upper airway-preferring respiratory stimulant with arousal properties, traits that have potential favorable relevance to some respiratory disorders but not others.NEW & NOTEWORTHY One of the major goals for translational sleep science and medicine is to identify viable and tractable pharmacological targets for obstructive sleep apnea and other respiratory disorders of sleep or sedation. In the present preclinical study in rats, we performed a randomized, within-subject, repeated-measures design over six intervention study days in chronically instrumented male and female rats with systemic peripheral administration of vehicle controls, the thyrotropin-releasing hormone analog taltirelin at two doses, all with and without coadministered trazodone. Trazodone was included due to clinical interest in the context of sleep apnea pharmacotherapy as it can suppress arousal without compromising pharyngeal muscle activity. These preclinical findings newly identify taltirelin as a stable upper airway-preferring respiratory stimulant with arousal properties. These traits have potential favorable relevance to some respiratory disorders but not others, as identified and discussed.


Asunto(s)
Fármacos del Sistema Respiratorio , Apnea Obstructiva del Sueño , Trazodona , Masculino , Femenino , Ratas , Animales , Hormona Liberadora de Tirotropina/farmacología , Hormona Liberadora de Tirotropina/uso terapéutico , Trazodona/farmacología , Trazodona/uso terapéutico , Fármacos del Sistema Respiratorio/farmacología , Fármacos del Sistema Respiratorio/uso terapéutico , Nivel de Alerta , Sueño/fisiología
6.
Neuroscience ; 468: 110-122, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34126184

RESUMEN

Alterations in thalamic GABAergic signaling are implicated in mediating the rise in 12-30 Hz electroencephalogram (EEG) activity that signals anesthetic-induced loss-of-consciousness with GABAA receptor-targeting general anesthetics. A number of modeling studies have identified that anesthetic-induced alterations in thalamocortico-corticothalamic signaling in the same network that generates sleep spindles would be sufficient to elicit this key EEG signature of anesthetic hypnosis with general anesthetic agents. Accordingly, we hypothesize that targeted stimulation of this thalamic GABAergic circuitry into a sleep-spindle mode of activity would promote the general anesthetic effects of etomidate. We recorded EEG activity and loss-of-righting reflex in transgenic mice expressing channel rhodopsin-2 on GABAergic neurons (ChR2-VGAT, n = 8) and control, wild-type mice (C57BL/6J, n = 8). On two consecutive days mice were randomly assigned to receive spindle-rhythm stimulation via an optical probe targeting the left reticular thalamic nucleus or no stimulation. After an initial 30-minute recording, mice were administered etomidate (12 mg/kg, intraperitoneal) and recorded for 90 min with or without optical stimulation. Etomidate elicited an increase in 12-30 Hz EEG power in wild-type and ChR2-VGAT mice for 20 min following administration (p < 0.001). Optical spindle-rhythm stimulation prolonged the increase in 12-30 Hz activity in ChR2-VGAT mice only (p = 0.023). Spindle-rhythm stimulation also increased the incidence and duration of sleep spindle-like oscillations in ChR2-VGAT mice only (all p ≤ 0.001). Despite the maintained anesthetic-like changes in EEG activity, optical spindle-rhythm stimulation was not associated with changes in the time to and duration of the loss-of-righting reflex, a behavioral endpoint of etomidate-induced general anesthesia in rodents.


Asunto(s)
Estado de Conciencia , Tálamo , Anestesia General , Animales , Electroencefalografía , Ratones , Ratones Endogámicos C57BL , Sueño , Inconsciencia/inducido químicamente
7.
Sleep ; 44(1)2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-32745213

RESUMEN

Obstructive sleep apnea (OSA) occurs exclusively during sleep due to reduced tongue motor activity. Withdrawal of excitatory inputs to the hypoglossal motor nucleus (HMN) from wake to sleep contributes to this reduced activity. Several awake-active neurotransmitters with inputs to the HMN (e.g. serotonin [5-HT]) inhibit K+ leak mediated by TASK-1/3 channels on hypoglossal motoneurons, leading to increased neuronal activity in vitro. We hypothesize that TASK channel inhibition at the HMN will increase tongue muscle activity in vivo and modulate responses to 5-HT. We first microperfused the HMN of anesthetized rats with TASK channel inhibitors: doxapram (75 µM, n = 9), A1899 (25 µM, n = 9), ML365 (25 µM, n = 9), acidified artificial cerebrospinal fluid (ACSF, pH = 6.25, n = 9); and a TASK channel activator terbinafine (50 µM, n = 9); all with and without co-applied 5-HT (10 mM). 5-HT alone at the HMN increased tongue motor activity (202.8% ± 45.9%, p < 0.001). However, neither the TASK channel inhibitors, nor activator, at the HMN changed baseline tongue activity (p > 0.716) or responses to 5-HT (p > 0.127). Tonic tongue motor responses to 5-HT at the HMN were also not different (p > 0.05) between ChAT-Cre:TASKf/f mice (n = 8) lacking TASK-1/3 channels on cholinergic neurons versus controls (n = 10). In freely behaving rats (n = 9), microperfusion of A1899 into the HMN increased within-breath phasic tongue motor activity in wakefulness only (p = 0.005) but not sleep, with no effects on tonic activity across all sleep-wake states. Together, the findings suggest robust maintenance of tongue motor activity despite various strategies for TASK channel manipulation targeting the HMN in vivo, and thus currently do not support this target and direction for potential OSA pharmacotherapy.


Asunto(s)
Nervio Hipogloso , Apnea Obstructiva del Sueño , Animales , Ratones , Neuronas Motoras , Ratas , Ratas Wistar , Apnea Obstructiva del Sueño/tratamiento farmacológico , Lengua
8.
eNeuro ; 7(6)2020.
Artículo en Inglés | MEDLINE | ID: mdl-33055199

RESUMEN

Neurons of the ventrolateral periaqueductal gray (vlPAG) and adjacent deep mesencephalic reticular nucleus (DpMe) are implicated in the control of sleep-wake state and are hypothesized components of a flip-flop circuit that maintains sleep bistability by preventing the overexpression of non-rapid eye movement (NREM)/REM sleep intermediary states (NRt). To determine the contribution of vlPAG/DpMe neurons in maintaining sleep bistability we combined computer simulations of flip-flop circuitry with focal inactivation of vlPAG/DpMe neurons by microdialysis delivery of the GABAA receptor agonist muscimol in freely behaving male rats (n = 25) instrumented for electroencephalographic and electromyographic recording. REM sleep was enhanced by muscimol at the vlPAG/DpMe, consistent with previous studies; however, our analyses of NRt dynamics in vivo and those produced by flop-flop circuit simulations show that current thinking is too narrowly focused on the contribution of REM sleep-inactive populations toward vlPAG/DpMe involvement in REM sleep control. We found that much of the muscimol-mediated increase in REM sleep was more appropriately classified as NRt. This loss of sleep bistability was accompanied by fragmentation of REM sleep, as evidenced by an increased number of short REM sleep bouts. REM sleep fragmentation stemmed from an increased number and duration of NRt bouts originating in REM sleep. By contrast, NREM sleep bouts were not likewise fragmented by vlPAG/DpMe inactivation. In flip-flop circuit simulations, these changes could not be replicated through inhibition of the REM sleep-inactive population alone. Instead, combined suppression of REM sleep active and inactive vlPAG/DpMe subpopulations was required to replicate the changes in NRt dynamics.


Asunto(s)
Sustancia Gris Periacueductal , Sueño , Animales , Electroencefalografía , Masculino , Neuronas , Ratas , Sueño REM , Vigilia
9.
Sleep ; 43(9)2020 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-32227104

RESUMEN

Thyrotropin-releasing hormone (TRH) is produced by the hypothalamus but most brain TRH is located elsewhere where it acts as a neuromodulator. TRH-positive neurons project to the hypoglossal motoneuron pool where TRH receptor RNA shows a high degree of differential expression compared with the rest of the brain. Strategies to modulate hypoglossal motor activity are of physiological and clinical interest given the potential for pharmacotherapy for obstructive sleep apnea (OSA), a common and serious respiratory disorder. Here, we identified the effects on tongue motor activity of TRH and a specific analog (taltirelin) applied locally to the hypoglossal motoneuron pool and systemically in vivo. Studies were performed under isoflurane anesthesia and across sleep-wake states in rats. In anesthetized rats, microperfusion of TRH (n = 8) or taltirelin (n = 9) into the hypoglossal motoneuron pool caused dose-dependent increases in tonic and phasic tongue motor activity (both p < 0.001). However, the motor responses to TRH were biphasic, being significantly larger "early" in the response versus at the end of the intervention (p ≤ 0.022). In contrast, responses to taltirelin were similar "early" versus "late" (p ≥ 0.107); i.e. once elicited, the motor responses to taltirelin were sustained and maintained. In freely behaving conscious rats (n = 10), microperfusion of 10 µM taltirelin into the hypoglossal motoneuron pool increased tonic and phasic tongue motor activity in non-rapid-eye-movement (REM) sleep (p ≤ 0.038). Intraperitoneal injection of taltirelin (1 mg/kg, n = 16 rats) also increased tonic tongue motor activity across sleep-wake states (p = 0.010). These findings inform the studies in humans to identify the potential beneficial effects of taltirelin for breathing during sleep and OSA.


Asunto(s)
Apnea Obstructiva del Sueño , Hormona Liberadora de Tirotropina , Animales , Nervio Hipogloso , Neuronas Motoras , Ratas , Sueño , Lengua
10.
Sci Rep ; 10(1): 550, 2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31953471

RESUMEN

Motoneurons are the final output pathway for the brain's influence on behavior. Here we identify properties of hypoglossal motor output to the tongue musculature. Tongue motor control is critical to the pathogenesis of obstructive sleep apnea, a common and serious sleep-related breathing disorder. Studies were performed on mice expressing a light sensitive cation channel exclusively on cholinergic neurons (ChAT-ChR2(H134R)-EYFP). Discrete photostimulations under isoflurane-induced anesthesia from an optical probe positioned above the medullary surface and hypoglossal motor nucleus elicited discrete increases in tongue motor output, with the magnitude of responses dependent on stimulation power (P < 0.001, n = 7) and frequency (P = 0.002, n = 8, with responses to 10 Hz stimulation greater than for 15-25 Hz, P < 0.022). Stimulations during REM sleep elicited significantly reduced responses at powers 3-20 mW compared to non-rapid eye movement (non-REM) sleep and wakefulness (each P < 0.05, n = 7). Response thresholds were also greater in REM sleep (10 mW) compared to non-REM and waking (3 to 5 mW, P < 0.05), and the slopes of the regressions between input photostimulation powers and output motor responses were specifically reduced in REM sleep (P < 0.001). This study identifies that variations in photostimulation input produce tunable changes in hypoglossal motor output in-vivo and identifies REM sleep specific suppression of net motor excitability and responsivity.


Asunto(s)
Channelrhodopsins/genética , Colina O-Acetiltransferasa/genética , Nervio Hipogloso/fisiología , Neuronas Motoras/fisiología , Lengua/inervación , Animales , Proteínas Bacterianas/genética , Isoflurano/administración & dosificación , Proteínas Luminiscentes/genética , Masculino , Ratones , Ratones Transgénicos , Sueño REM , Lengua/fisiología , Vigilia/fisiología
11.
Sci Rep ; 9(1): 14122, 2019 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-31575947

RESUMEN

Opioid drugs are the mainstay of pain management but present the side-effect of respiratory depression that can be lethal with overdose. In addition to their respiratory effect, opioids also induce a profound sedative state and produce electrocortical features characteristic of a state of reduced brain arousal, similar to anaesthesia or sleep. In such states, respiratory activity depends more on the integrity of the brainstem respiratory network than it does during wakefulness. Accordingly, we propose that sedation by fentanyl induces specific electrocortical changes consistent with reduced brain arousal, and that the magnitude of respiratory depression is associated with distinct electrocortical changes. To these aims, we determined the effects of systemic injections of fentanyl (dosage 100 µg ·kg) versus control on electrocortical  and respiratory activities of freely-behaving rats. We found that fentanyl induced electrocortical changes that differed from those observed in sleep or wakefulness. Fentanyl increased δ (1-3 Hz) frequency power (P < 0.001), but reduced α (7.5-13.5 Hz) and ß2 (20-30 Hz) powers (P = 0.012 and P < 0.001, respectively), when compared to wakefulness. Interestingly, respiratory rate depression by fentanyl was significantly correlated with increased θ power (R = 0.61, P < 0.001), therefore showing a clear association between electrocortical activity and the magnitude of respiratory rate depression. Overall, we provide new evidence linking specific electrocortical changes to the severity of respiratory depression by opioids, which highlights the importance of considering the cortical and subcortical effects of opioids in addition to their impacts on breathing when evaluating opioid-induced respiratory depression.


Asunto(s)
Analgésicos Opioides/farmacología , Fentanilo/farmacología , Insuficiencia Respiratoria/tratamiento farmacológico , Frecuencia Respiratoria/efectos de los fármacos , Anestesia/métodos , Animales , Nivel de Alerta/efectos de los fármacos , Tronco Encefálico/efectos de los fármacos , Masculino , Ratas , Ratas Wistar
12.
Sleep ; 41(8)2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29741674

RESUMEN

Study Objectives: Previous research has suggested that general anesthetics can disturb postoperative sleep patterns by affecting the sleep-wake cycle. The objective was to identify the effects of general anesthetics on sleep quality and related behavioral changes in children. Methods: This was a prospective, observational case-control study with children, aged 18 months to 8 years, undergoing general anesthesia for elective surgery. Participants wore an actigraph for 7 days on three occasions: prior to surgery, the immediate postoperative period, and 3 months after surgery. Data regarding behavior patterns were collected using behavioral assessments at baseline, the first postoperative week, and 3 months following surgery. Results: Thirty-one participants (mean age 4.8 ± 2.0 years, 81% male) underwent urologic or otolaryngologic surgery. The median (interquartile range) anesthetic duration was 132.0 (80.0-184.0) min. No significant differences were found in sleep efficiency, total sleep time, wake time after sleep onset, or sleep onset latency between baseline, 7 day postoperative period, and the 3 month follow-up. No significant differences were found in sleep-related behavioral metrics including internalizing and externalizing behaviors, and executive functioning. Data were compared with a control group of 18 participants (mean age 5.3 ± 1.8 years, 61% male). No significant differences were found in sleep patterns and related behavioral metrics between both groups. Conclusions: In this study, general anesthesia did not result in disturbed sleep or associated negative behavioral changes in otherwise healthy children undergoing elective surgeries of low complexity. Physicians can advise parents that a child's surgery and associated general anesthetic exposure may not result in significant changes in postoperative sleep patterns.


Asunto(s)
Anestesia General/efectos adversos , Trastornos del Sueño-Vigilia/inducido químicamente , Sueño/efectos de los fármacos , Actigrafía , Estudios de Casos y Controles , Niño , Preescolar , Procedimientos Quirúrgicos Electivos , Femenino , Humanos , Lactante , Masculino , Padres , Periodo Posoperatorio , Estudios Prospectivos , Sueño/fisiología
13.
Sci Rep ; 7(1): 18105, 2017 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-29273726

RESUMEN

Persistent and stable respiratory activity across behavioral states is key to homeostasis. Extrasynaptic δ-subunit containing GABAA receptors (δGABAARs) mediate tonic inhibition and regulate network activity. However, the influence of δGABAARs on respiratory rhythm and motor outputs is unknown. We manipulated extra-synaptic GABAA receptor function in the preBötzinger Complex (preBötC), a site central to the generation of inspiratory motor activity in mammals. Activation of preBötC δGABAARs in anesthetized rats and wild-type mice decreased breathing rate. In δGABAAR knockout (Gabrd -/-) mice, however, δGABAARs activation had no effect on breathing rate. We then found that during active wakefulness associated with behaviors and movements, diaphragm activation was higher in the Gabrd -/- compared to wild-type mice, but not in other states. These findings identify that δGABAARs modulate the respiratory network, which is critical to understand how δGABAARs change breathing in pathological conditions affecting extra-synaptic GABAA receptor function such as exposure to anesthetics and neurosteroids.


Asunto(s)
Bulbo Raquídeo/fisiología , Músculos del Cuello/fisiología , Neuronas/fisiología , Receptores de GABA-A/metabolismo , Frecuencia Respiratoria/fisiología , Animales , Conducta Animal/fisiología , Ratones , Ratones Noqueados , Ratas
14.
Continuum (Minneap Minn) ; 23(4, Sleep Neurology): 955-972, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28777170

RESUMEN

PURPOSE OF REVIEW: This article outlines the fundamental brain mechanisms that control sleep-wake patterns and reviews how pathologic changes in these control mechanisms contribute to common sleep disorders. RECENT FINDINGS: Discrete but interconnected clusters of cells located within the brainstem and hypothalamus comprise the circuits that generate wakefulness, non-rapid eye movement (non-REM) sleep, and REM sleep. These clusters of cells use specific neurotransmitters, or collections of neurotransmitters, to inhibit or excite their respective sleep- and wake-promoting target sites. These excitatory and inhibitory connections modulate not only the presence of wakefulness or sleep, but also the levels of arousal within those states, including the depth of sleep, degree of vigilance, and motor activity. Dysfunction or degeneration of wake- and sleep-promoting circuits is associated with narcolepsy, REM sleep behavior disorder, and age-related sleep disturbances. SUMMARY: Research has made significant headway in identifying the brain circuits that control wakefulness, non-REM, and REM sleep and has led to a deeper understanding of common sleep disorders and disturbances.


Asunto(s)
Encéfalo/fisiopatología , Trastorno de la Conducta del Sueño REM/terapia , Sueño/fisiología , Vigilia/fisiología , Animales , Encéfalo/fisiología , Humanos , Narcolepsia/fisiopatología , Neuronas/fisiología , Trastorno de la Conducta del Sueño REM/diagnóstico
15.
Respirology ; 22(5): 861-873, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28544082

RESUMEN

There is currently no pharmacotherapy for obstructive sleep apnoea (OSA) but there is no principled a priori reason why there should not be one. This review identifies a rational decision-making strategy with the necessary logical underpinnings that any reasonable approach would be expected to navigate to develop a viable pharmacotherapy for OSA. The process first involves phenotyping an individual to quantify and characterize the critical predisposing factor(s) to their OSA pathogenesis and identify, a priori, if the patient is likely to benefit from a pharmacotherapy that targets those factors. We then identify rational strategies to manipulate those critical predisposing factor(s), and the barriers that have to be overcome for success of any OSA pharmacotherapy. A new analysis then identifies candidate drug targets to manipulate the upper airway motor circuitry for OSA pharmacotherapy. The first conclusion is that there are two general pharmacological approaches for OSA treatment that are of the most potential benefit and are practically realistic, one being fairly intuitive but the second perhaps less so. The second conclusion is that after identifying the critical physiological obstacles to OSA pharmacotherapy, there are current therapeutic targets of high interest for future development. The final analysis provides a tabulated resource of 'druggable' targets that are relatively restricted to the circuitry controlling the upper airway musculature, with these candidate targets being of high priority for screening and further study. We also emphasize that a pharmacotherapy may not cure OSA per se, but may still be a useful adjunct to improve the effectiveness of, and adherence to, other treatment mainstays.


Asunto(s)
Toma de Decisiones , Descubrimiento de Drogas , Apnea Obstructiva del Sueño/tratamiento farmacológico , Humanos , Fenotipo , Apnea Obstructiva del Sueño/fisiopatología
16.
Sci Rep ; 7: 45860, 2017 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-28383527

RESUMEN

Reduced tongue muscle tone precipitates obstructive sleep apnea (OSA), and activation of the tongue musculature can lessen OSA. The hypoglossal motor nucleus (HMN) innervates the tongue muscles but there is no pharmacological agent currently able to selectively manipulate a channel (e.g., Kir2.4) that is highly restricted in its expression to cranial motor pools such as the HMN. To model the effect of manipulating such a restricted target, we introduced a "designer" receptor into the HMN and selectively modulated it with a "designer" drug. We used cre-dependent viral vectors (AAV8-hSyn-DIO-hM3Dq-mCherry) to transduce hypoglossal motoneurons of ChAT-Cre+ mice with hM3Dq (activating) receptors. We measured sleep and breathing in three conditions: (i) sham, (ii) after systemic administration of clozapine-N-oxide (CNO; 1 mg/kg) or (iii) vehicle. CNO activates hM3Dq receptors but is otherwise biologically inert. Systemic administration of CNO caused significant and sustained increases in tongue muscle activity in non-REM (261 ± 33% for 10 hrs) and REM sleep (217 ± 21% for 8 hrs), both P < 0.01 versus controls. Responses were specific and selective for the tongue with no effects on diaphragm or postural muscle activities, or sleep-wake states. These results support targeting a selective and restricted "druggable" target at the HMN (e.g., Kir2.4) to activate tongue motor activity during sleep.


Asunto(s)
Apnea Obstructiva del Sueño/fisiopatología , Sueño/fisiología , Lengua/fisiología , Animales , Clozapina/administración & dosificación , Clozapina/análogos & derivados , Diafragma/inervación , Diafragma/fisiología , Vías Eferentes/efectos de los fármacos , Vías Eferentes/fisiología , Electromiografía , Músculos Faciales/inervación , Músculos Faciales/fisiología , Nervio Hipogloso/fisiología , Neuronas Motoras/fisiología , Ratas , Ratas Wistar , Serotonina/metabolismo , Sueño/efectos de los fármacos , Apnea Obstructiva del Sueño/tratamiento farmacológico , Lengua/efectos de los fármacos , Lengua/inervación , Vigilia/efectos de los fármacos , Vigilia/fisiología
17.
Mol Brain ; 10(1): 10, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-28385162

RESUMEN

Human studies demonstrate that sleep impairment is a concurrent comorbidity of autism spectrum disorders (ASD), but its etiology remains largely uncertain. One of the prominent theories of ASD suggests that an imbalance in synaptic excitation/inhibition may contribute to various aspects of ASD, including sleep impairments. Following the identification of Nlgn3R451C mutation in patients with ASD, its effects on synaptic transmission and social behaviours have been examined extensively in the mouse model. However, the contributory role of this mutation to sleep impairments in ASD remains unknown. In this study, we showed that Nlgn3R451C knock-in mice, an established genetic model for ASD, exhibited normal duration and distribution of sleep/wake states but significantly altered electroencephalography (EEG) power spectral profiles for wake and sleep.


Asunto(s)
Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/fisiopatología , Moléculas de Adhesión Celular Neuronal/genética , Electroencefalografía , Proteínas de la Membrana/genética , Mutación/genética , Proteínas del Tejido Nervioso/genética , Animales , Modelos Animales de Enfermedad , Electromiografía , Masculino , Ratones Mutantes , Sueño REM/fisiología , Factores de Tiempo , Vigilia/fisiología
18.
J Neurosci ; 37(15): 4007-4022, 2017 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-28209737

RESUMEN

Cataplexy is a hallmark of narcolepsy characterized by the sudden uncontrollable onset of muscle weakness or paralysis during wakefulness. It can occur spontaneously, but is typically triggered by positive emotions such as laughter. Although cataplexy was identified >130 years ago, its neural mechanism remains unclear. Here, we show that a newly identified GABA circuit within the central nucleus of the amygdala (CeA) promotes cataplexy. We used behavioral, electrophysiological, immunohistochemical, and chemogenetic strategies to target and manipulate CeA activity selectively in narcoleptic (orexin-/-) mice to determine its functional role in controlling cataplexy. First, we show that chemogenetic activation of the entire CeA produces a marked increase in cataplexy attacks. Then, we show that GABA cells within the CeA are responsible for mediating this effect. To manipulate GABA cells specifically, we developed a new mouse line that enables genetic targeting of GABA cells in orexin-/- mice. We found that chemogenetic activation of GABA CeA cells triggered a 253% increase in the number of cataplexy attacks without affecting their duration, suggesting that GABA cells play a functional role in initiating but not maintaining cataplexy. We show that GABA cell activation only promotes cataplexy attacks associated with emotionally rewarding stimuli, not those occurring spontaneously. However, we found that chemogenetic inhibition of GABA CeA cells does not prevent cataplexy, suggesting these cells are not required for initiating cataplexy attacks. Our results indicate that the CeA promotes cataplexy onset and that emotionally rewarding stimuli may trigger cataplexy by activating GABA cells in the CeA.SIGNIFICANCE STATEMENT Although cataplexy has been closely linked to positive emotions for >130 years, the neural circuitry that underlies this relationship is poorly understood. Recent work suggests that the amygdala, a brain area important for processing emotion, may be part of this circuit. This study provides the first functional evidence to implicate GABA cells in the amygdala as regulators of cataplexy triggered by positive emotions and identifies the amygdala as the brain region important more for gating the entrance into rather than the exit from cataplexy. We also generated a new mouse model for studying GABA neurons in narcoleptic mice, which could serve as a useful tool for studying the neurobiological underpinnings of narcolepsy.


Asunto(s)
Cataplejía/metabolismo , Núcleo Amigdalino Central/metabolismo , Neuronas GABAérgicas/metabolismo , Animales , Cataplejía/fisiopatología , Núcleo Amigdalino Central/fisiopatología , Electroencefalografía/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Orexinas/deficiencia
19.
Neurotoxicol Teratol ; 61: 115-122, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27793660

RESUMEN

A reduction in the activity of GABAA receptors, particularly α5 subunit-containing GABAA receptors (α5GABAARs), has been implicated in the etiology of Autism Spectrum Disorders (ASD). Genetically modified mice that lack α5GABAARs (Gabra5-/-) exhibit autism-like behaviors and both enhanced and impaired learning and memory, depending on the behavioral task. The aim of this study was to examine the electroencephalogram (EEG) activity and sleep-wake behaviors in Gabra5-/- mice and wild-type mice. In addition, since some individuals with ASD can exhibit elevated innate immune response, mice were treated with lipopolysaccharide (LPS; 125mg/kg intraperitoneal injection) or vehicle and EEG and sleep-wake patterns were assessed. The results showed that Gabra5-/- mice (n=3) exhibited elevated 0-2Hz EEG activity during all sleep-wake states (all p<0.04), decreased 8-12Hz EEG activity during REM sleep (p=0.04), and decreased sleep spindles under baseline conditions compared to wild-type controls (n=4) (all p≤0.03). Alterations in EEG activity and sleep-wake behavior were identified in Gabra5-/- mice following treatment with LPS, however these changes were similar to those in wild-type mice. Our findings support the hypothesis that reduced α5GABAAR activity contributes to an ASD phenotype. The results also suggest that Gabra5-/- mice may serve as an animal model for ASD, as assessed through EEG activity and sleep-wake behaviors.


Asunto(s)
Trastorno del Espectro Autista/fisiopatología , Receptores de GABA-A/fisiología , Fases del Sueño/fisiología , Vigilia/fisiología , Animales , Trastorno del Espectro Autista/genética , Modelos Animales de Enfermedad , Electroencefalografía/efectos de los fármacos , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Noqueados , Receptores de GABA-A/biosíntesis , Receptores de GABA-A/deficiencia , Receptores de GABA-A/genética , Fases del Sueño/efectos de los fármacos , Fases del Sueño/genética , Vigilia/genética
20.
Sci Rep ; 6: 32707, 2016 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-27599866

RESUMEN

Breathing is generated by a respiratory network in the brainstem. At its core, a population of neurons expressing neurokinin-1 receptors (NK1R) and the peptide somatostatin (SST) form the preBötzinger Complex (preBötC), a site essential for the generation of breathing. PreBötC interneurons generate rhythm and follower neurons shape motor outputs by activating upper airway respiratory muscles. Since NK1R-expressing preBötC neurons are preferentially inhibited by µ-opioid receptors via activation of GIRK channels, NK1R stimulation may also involve GIRK channels. Hence, we identify the contribution of GIRK channels to rhythm, motor output and respiratory modulation by NK1Rs and SST. In adult rats, GIRK channels were identified in NK1R-expressing preBötC cells. Their activation decreased breathing rate and genioglossus muscle activity, an important upper airway muscle. NK1R activation increased rhythmic breathing and genioglossus muscle activity in wild-type mice, but not in mice lacking GIRK2 subunits (GIRK2(-/-)). Conversely, SST decreased rhythmic breathing via SST2 receptors, reduced genioglossus muscle activity likely through SST4 receptors, but did not involve GIRK channels. In summary, NK1R stimulation of rhythm and motor output involved GIRK channels, whereas SST inhibited rhythm and motor output via two SST receptor subtypes, therefore revealing separate circuits mediating rhythm and motor output.


Asunto(s)
Tronco Encefálico/fisiología , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/fisiología , Receptores de Neuroquinina-1/fisiología , Somatostatina/fisiología , Animales , Inmunohistoquímica , Masculino , Ratones , Ratas , Ratas Wistar , Respiración
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...