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

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neurosci ; 37(15): 4007-4022, 2017 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-28209737

RESUMO

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.


Assuntos
Cataplexia/metabolismo , Núcleo Central da Amígdala/metabolismo , Neurônios GABAérgicos/metabolismo , Animais , Cataplexia/fisiopatologia , Núcleo Central da Amígdala/fisiopatologia , Eletroencefalografia/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Orexinas/deficiência
2.
Respirology ; 22(5): 861-873, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28544082

RESUMO

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.


Assuntos
Tomada de Decisões , Descoberta de Drogas , Apneia Obstrutiva do Sono/tratamento farmacológico , Humanos , Fenótipo , Apneia Obstrutiva do Sono/fisiopatologia
3.
J Neurosci ; 35(40): 13555-67, 2015 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-26446210

RESUMO

Basal forebrain cholinergic neurons are the main source of cortical acetylcholine, and their activation by histamine elicits cortical arousal. TWIK-like acid-sensitive K(+) (TASK) channels modulate neuronal excitability and are expressed on basal forebrain cholinergic neurons, but the role of TASK channels in the histamine-basal forebrain cholinergic arousal circuit is unknown. We first expressed TASK channel subunits and histamine Type 1 receptors in HEK cells. Application of histamine in vitro inhibited the acid-sensitive K(+) current, indicating a functionally coupled signaling mechanism. We then studied the role of TASK channels in modulating electrocortical activity in vivo using freely behaving wild-type (n = 12) and ChAT-Cre:TASK(f/f) mice (n = 12), the latter lacking TASK-1/3 channels on cholinergic neurons. TASK channel deletion on cholinergic neurons significantly altered endogenous electroencephalogram oscillations in multiple frequency bands. We then identified the effect of TASK channel deletion during microperfusion of histamine into the basal forebrain. In non-rapid eye movement sleep, TASK channel deletion on cholinergic neurons significantly attenuated the histamine-induced increase in 30-50 Hz activity, consistent with TASK channels contributing to histamine action on basal forebrain cholinergic neurons. In contrast, during active wakefulness, histamine significantly increased 30-50 Hz activity in ChAT-Cre:TASK(f/f) mice but not wild-type mice, showing that the histamine response depended upon the prevailing cortical arousal state. In summary, we identify TASK channel modulation in response to histamine receptor activation in vitro, as well as a role of TASK channels on cholinergic neurons in modulating endogenous oscillations in the electroencephalogram and the electrocortical response to histamine at the basal forebrain in vivo. SIGNIFICANCE STATEMENT: Attentive states and cognitive function are associated with the generation of γ EEG activity. Basal forebrain cholinergic neurons are important modulators of cortical arousal and γ activity, and in this study we investigated the mechanism by which these neurons are activated by the wake-active neurotransmitter histamine. We found that histamine inhibited a class of K(+) leak channels called TASK channels and that deletion of TASK channels selectively on cholinergic neurons modulated baseline EEG activity as well as histamine-induced changes in γ activity. By identifying a discrete brain circuit where TASK channels can influence γ activity, these results represent new knowledge that enhances our understanding of how subcortical arousal systems may contribute to the generation of attentive states.


Assuntos
Nível de Alerta/efeitos dos fármacos , Prosencéfalo Basal/citologia , Córtex Cerebral/fisiologia , Neurônios Colinérgicos/efeitos dos fármacos , Agonistas dos Receptores Histamínicos/farmacologia , Histamina/farmacologia , Proteínas do Tecido Nervoso/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Animais , Prosencéfalo Basal/fisiologia , Córtex Cerebral/efeitos dos fármacos , Colina O-Acetiltransferase/metabolismo , Eletroencefalografia , Eletromiografia , Ritmo Gama/efeitos dos fármacos , Ritmo Gama/genética , Humanos , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Lectinas de Plantas/metabolismo , Canais de Potássio de Domínios Poros em Tandem/genética , Sono
4.
Anesthesiology ; 125(5): 964-978, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27552653

RESUMO

BACKGROUND: Alterations in thalamic γ-aminobutyric acid-mediated signaling are thought to underlie the increased frontal α-ß frequency electrocortical activity that signals anesthetic-induced loss of consciousness with γ-aminobutyric acid receptor type A (GABAAR)-targeting general anesthetics. The general anesthetic etomidate elicits phasic extrasynaptic GABAAR activation ("spillover" inhibition) at thalamocortical neurons in vitro. We hypothesize that this action of etomidate at the thalamus is sufficient to trigger an increase in frontal α-ß frequency electrocortical activity and that this effect of etomidate is fully recapitulated by enhanced thalamic spillover inhibition in vivo. METHODS: We recorded electrocortical activity and sleep-wake behavior in freely behaving wild-type (n = 33) and extrasynaptic δ-subunit-containing GABAAR knockout mice (n = 9) during bilateral microperfusion of the thalamus with etomidate and/or other pharmacologic agents that influence GABAAR or T-type Ca channel activity. RESULTS: Microperfusion of etomidate into the thalamus elicited an increase in α-ß frequency electrocortical activity that occurred only during non-rapid-eye-movement (REM) sleep (11.0 ± 11.8% and 16.0 ± 14.2% greater 8 to 12- and 12 to 30-Hz power, respectively; mean ± SD; both P < 0.031) and was not affected by blockade of thalamic T-type Ca channels. Etomidate at the thalamus also increased spindle-like oscillations during non-REM sleep (4.5 ± 2.4 spindle per minute with etomidate vs. 3.2 ± 1.7 at baseline; P = 0.002). These effects of etomidate were fully recapitulated by enhanced thalamic extrasynaptic GABAAR-mediated spillover inhibition. CONCLUSIONS: These findings identify how a prototypic GABAAR-targeting general anesthetic agent can elicit the characteristic brain wave pattern associated with anesthetic hypnosis when acting at the thalamus by promoting spillover inhibition and the necessity of a preexisting non-REM mode of activity in the thalamus to generate this effect.


Assuntos
Etomidato/farmacologia , Hipnóticos e Sedativos/farmacologia , Sono/efeitos dos fármacos , Tálamo/efeitos dos fármacos , Animais , Eletroencefalografia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos
5.
Anesthesiology ; 124(3): 641-50, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26675532

RESUMO

BACKGROUND: Drugs acting on µ-opioid receptors (MORs) are widely used as analgesics but present side effects including life-threatening respiratory depression. MORs are G-protein-coupled receptors inhibiting neuronal activity through calcium channels, adenylyl cyclase, and/or G-protein-gated inwardly rectifying potassium (GIRK) channels. The pathways underlying MOR-dependent inhibition of rhythmic breathing are unknown. METHODS: By using a combination of genetic, pharmacological, and physiological tools in rodents in vivo, the authors aimed to identify the role of GIRK channels in MOR-mediated inhibition of respiratory circuits. RESULTS: GIRK channels were expressed in the ventrolateral medulla, a neuronal population regulating rhythmic breathing, and GIRK channel activation with flupirtine reduced respiratory rate in rats (percentage of baseline rate in mean ± SD: 79.4 ± 7.4%, n = 7), wild-type mice (82.6 ± 3.8%, n = 3), but not in mice lacking the GIRK2 subunit, an integral subunit of neuronal GIRK channels (GIRK2, 101.0 ± 1.9%, n = 3). Application of the MOR agonist [D-Ala, N-MePhe, Gly-ol]-enkephalin (DAMGO) to the ventrolateral medulla depressed respiratory rate, an effect partially reversed by the GIRK channel blocker Tertiapin-Q (baseline: 42.1 ± 7.4 breath/min, DAMGO: 26.1 ± 13.4 breath/min, Tertiapin-Q + DAMGO: 33.9 ± 9.8 breath/min, n = 4). Importantly, DAMGO applied to the ventrolateral medulla failed to reduce rhythmic breathing in GIRK2 mice (percentage of baseline rate: 103.2 ± 12.1%, n = 4), whereas it considerably reduced rate in wild-type mice (62.5 ± 17.7% of baseline, n = 4). Respiratory rate depression by systemic injection of the opioid analgesic fentanyl was markedly reduced in GIRK2 (percentage of baseline: 12.8 ± 15.8%, n = 5) compared with wild-type mice (72.9 ± 27.3%). CONCLUSIONS: Overall, these results identify that GIRK channels contribute to respiratory inhibition by MOR, an essential step toward understanding respiratory depression by opioids.


Assuntos
Analgésicos Opioides/toxicidade , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Insuficiência Respiratória/induzido quimicamente , Insuficiência Respiratória/metabolismo , Animais , Venenos de Abelha/farmacologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/toxicidade , Feminino , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/antagonistas & inibidores , Masculino , Camundongos , Camundongos Knockout , Ratos , Ratos Wistar , Receptores Opioides mu/agonistas , Receptores Opioides mu/fisiologia
6.
Anesthesiology ; 125(5): 889-903, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27541315

RESUMO

BACKGROUND: Opioid analgesia is an essential component of perioperative care, but effective analgesia can be limited by excessive sedation and respiratory depression. The cortical signatures associated with sedation by opioids and the relationship between changes in cortical activity and respiratory function are not well understood. The objectives of this study were to identify the electroencephalogram signatures of sedation and respiratory changes induced by morphine in a pediatric population after elective surgery. METHODS: After otologic surgery, patients (14.8 ± 2.8 yr, n = 10) stayed overnight for pain relief with morphine (3 to 10 mg), hydration, and clinical observation. Electroencephalogram activity and polysomnography were performed before and after morphine, and electroencephalogram spectral properties and cardiorespiratory activities were analyzed. RESULTS: Compared to wakefulness and non-rapid eye movement sleep, morphine reduced high-frequency ß1 (13.5 to 20 Hz) and ß2 (20 to 30Hz) electroencephalogram powers (n = 10) and decreased coherence between frontal and occipital ß2 electroencephalogram activities (n = 9), therefore indicating that morphine induced a deep sedative state. Morphine also reduced respiratory rate by 8.3% (n = 10). Interestingly, there was a significant correlation between the reduction in ß1 electroencephalogram activity and the depression in respiratory rate induced by morphine (R = 0.715, n = 10). With significant reduction in ß1 power, respiratory rate was decreased by more than 25%, suggesting that reduction in cortical arousal is associated with the severity of respiratory rate depression. CONCLUSIONS: Analgesic doses of morphine are associated with reduction in respiratory rate when accompanied by reduction in ß1 electroencephalogram power, indicating a powerful effect of cortical arousal state per se in respiratory rate depression by morphine.


Assuntos
Analgésicos Opioides/farmacologia , Encéfalo/efeitos dos fármacos , Estado de Consciência/efeitos dos fármacos , Morfina/farmacologia , Insuficiência Respiratória/fisiopatologia , Taxa Respiratória/efeitos dos fármacos , Adolescente , Criança , Pré-Escolar , Eletroencefalografia/efeitos dos fármacos , Feminino , Humanos , Masculino , Polissonografia/efeitos dos fármacos
7.
Anesthesiology ; 124(2): 417-27, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26566283

RESUMO

BACKGROUND: Critically ill patients with severe inflammation often exhibit heightened sensitivity to general anesthetics; however, the underlying mechanisms remain poorly understood. Inflammation increases the number of γ-aminobutyric acid type A (GABAA) receptors expressed on the surface of neurons, which supports the hypothesis that inflammation increases up-regulation of GABAA receptor activity by anesthetics, thereby enhancing the behavioral sensitivity to these drugs. METHODS: To mimic inflammation in vitro, cultured hippocampal and cortical neurons were pretreated with interleukin (IL)-1ß. Whole cell patch clamp methods were used to record currents evoked by γ-aminobutyric acid (GABA) (0.5 µM) in the absence and presence of etomidate or isoflurane. To mimic inflammation in vivo, mice were treated with lipopolysaccharide, and several anesthetic-related behavioral endpoints were examined. RESULTS: IL-1ß increased the amplitude of current evoked by GABA in combination with clinically relevant concentrations of either etomidate (3 µM) or isoflurane (250 µM) (n = 5 to 17, P < 0.05). Concentration-response plots for etomidate and isoflurane showed that IL-1ß increased the maximal current 3.3-fold (n = 5 to 9) and 1.5-fold (n = 8 to 11), respectively (P < 0.05 for both), whereas the half-maximal effective concentrations were unchanged. Lipopolysaccharide enhanced the hypnotic properties of both etomidate and isoflurane. The immobilizing properties of etomidate, but not isoflurane, were also increased by lipopolysaccharide. Both lipopolysaccharide and etomidate impaired contextual fear memory. CONCLUSIONS: These results provide proof-of-concept evidence that inflammation increases the sensitivity of neurons to general anesthetics. This increase in anesthetic up-regulation of GABAA receptor activity in vitro correlates with enhanced sensitivity for GABAA receptor-dependent behavioral endpoints in vivo.


Assuntos
Anestésicos Gerais/farmacologia , Inflamação/fisiopatologia , Neurônios/efeitos dos fármacos , Anestésicos Inalatórios/farmacologia , Animais , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Etomidato/farmacologia , Hipnóticos e Sedativos/farmacologia , Isoflurano/farmacologia , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Patch-Clamp , Receptores de GABA-A/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Ácido gama-Aminobutírico/efeitos dos fármacos
8.
J Neurosci ; 34(37): 12253-66, 2014 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-25209268

RESUMO

Extrasynaptic δ-subunits containing GABAA receptors (δGABAARs) are sensitive targets for several commonly used hypnotic agents and mediate tonic neuronal inhibition. δGABAARs are highly expressed within the thalamus and their activation promotes a switch from tonic to burst firing in vitro. Here we test two hypotheses in vivo. (1) Activation of thalamic δGABAARs will elicit electrocortical signatures consistent with widespread thalamocortical burst firing such as increased delta oscillations (1-4 Hz) and reciprocal changes in spindle-like oscillations (7-14 Hz). (2) These signatures will be recapitulated by the general anesthetic etomidate, if the electrocortical effects of etomidate at the thalamus are mediated by δGABAARs. Microperfusion of the δGABAAR-preferring agonist 4,5,6,7-tetrahydroisoxazolo[5,4-c]pyridin-3-ol (THIP; 10 and 50 µM) into the ventrobasal complex produced significant effects on electrocortical activity in wild-type mice, but not in mice lacking δGABAARs (Gabrd(-/-)), i.e., the effects with THIP were dependent on δGABAARs. THIP (1) increased 1-4 Hz power in wakefulness and nonrapid-eye movement (NREM) sleep; (2) reduced spindle-like oscillations in NREM sleep; and (3) increased the speed of stable transitions into NREM sleep, indicating effects on state-space dynamics. In contrast, microperfusion of etomidate (10 and 30 µM) into the ventrobasal complex produced effects on electrocortical activity that were independent of δGABAARs, i.e., effects occurred in wild-type and Gabrd(-/-) mice. Etomidate (1) decreased 1-4 Hz power, increased 8-12 Hz, and/or 12-30 Hz power in all sleep-wake states; (2) increased spindle-like oscillations; and (3) increased REM sleep expression. These results indicate that thalamic δGABAARs promote electrocortical signatures of deep NREM sleep, but do not mediate the effects of etomidate at the thalamus in vivo.


Assuntos
Potenciais de Ação/fisiologia , Relógios Biológicos/fisiologia , Etomidato/farmacologia , Rede Nervosa/fisiologia , Receptores de GABA-A/química , Receptores de GABA-A/metabolismo , Fases do Sono/fisiologia , Tálamo/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Relógios Biológicos/efeitos dos fármacos , Hipnóticos e Sedativos/farmacologia , Masculino , Camundongos , Camundongos Knockout , Rede Nervosa/efeitos dos fármacos , Subunidades Proteicas , Fases do Sono/efeitos dos fármacos , Tálamo/efeitos dos fármacos
9.
J Neurosci ; 34(43): 14198-209, 2014 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-25339734

RESUMO

Initial theories of rapid eye movement (REM) sleep generation posited that induction of the state required activation of the pontine subceruleus (SubC) by cholinergic inputs. Although the capacity of cholinergic neurotransmission to contribute to REM sleep generation has been established, the role of cholinergic inputs in the generation of REM sleep is ultimately undetermined as the critical test of this hypothesis (local blockade of SubC acetylcholine receptors) has not been rigorously performed. We used bilateral microdialysis in freely behaving rats (n = 32), instrumented for electroencephalographic and electromyographic recording, to locally manipulate neurotransmission in the SubC with select drugs. As predicted, combined microperfusion of D-AP5 (glutamate receptor antagonist) and muscimol (GABAA receptor agonist) in the SubC virtually eliminated REM sleep. However, REM sleep was not reduced by scopolamine microperfusion in this same region, at a concentration capable of blocking the effects of cholinergic receptor stimulation. This result suggests that transmission of REM sleep drive to the SubC is acetylcholine-independent. Although SubC cholinergic inputs are not majorly involved in REM sleep generation, they may perform a minor function in the reinforcement of transitions into REM sleep, as evidenced by increases in non-REM-to-REM sleep transition duration and failure rate during cholinergic receptor blockade. Cholinergic receptor antagonism also attenuated the normal increase in hippocampal θ oscillations that characterize REM sleep. Using computational modeling, we show that our in vivo results are consistent with a mutually excitatory interaction between the SubC and cholinergic neurons where, importantly, cholinergic neuron activation is gated by SubC activity.


Assuntos
Neurônios Colinérgicos/fisiologia , Ponte/fisiologia , Sono REM/fisiologia , Animais , Neurônios Colinérgicos/efeitos dos fármacos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Masculino , Rede Nervosa/efeitos dos fármacos , Rede Nervosa/fisiologia , Ponte/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores Colinérgicos/fisiologia , Sono REM/efeitos dos fármacos
10.
J Neurosci ; 33(20): 8716-28, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23678115

RESUMO

How rhythms are generated by neuronal networks is fundamental to understand rhythmic behaviors such as respiration, locomotion, and mastication. Respiratory rhythm is generated by the preBötzinger complex (preBötC), an anatomically and functionally discrete population of brainstem neurons, central and necessary for respiratory rhythm. In specific in vitro conditions, preBötC neurons depend on voltage-dependent inward currents to generate respiratory rhythm. In the mature and intact organism, where preBötC neurons are deeply embedded in the respiratory network, the contribution of ionic currents to respiratory rhythm is unclear. We propose that a set of ionic currents plays a key role in generating respiratory rhythm in the mature organism in vivo. By microperfusing ionic current blockers into the preBötC of adult rats, we identify the hyperpolarization-activated cation current as a critical component of the mechanism promoting respiratory rhythm, and that this current, in combination with the persistent sodium current, is essential to respiratory rhythm in vivo. Importantly, both currents contribute to rhythmic activity in states of anesthesia, quiet wakefulness, and sleep, but not when the organism is engaged in active behaviors. These data show that a set of ionic currents at the preBötC imparts the network with rhythmicity in reduced states of arousal, although the network can override their contribution to adjust its activity for nonrhythmic behaviors in active wakefulness.


Assuntos
Periodicidade , Centro Respiratório/fisiologia , Mecânica Respiratória/fisiologia , Canais de Sódio/fisiologia , Simportadores de Cloreto de Sódio-Potássio/fisiologia , Análise de Variância , Animais , Fármacos Cardiovasculares/farmacologia , Eletroencefalografia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Lateralidade Funcional/efeitos dos fármacos , Lateralidade Funcional/fisiologia , Técnicas In Vitro , Masculino , Potenciais da Membrana/efeitos dos fármacos , Microdiálise , Atividade Motora/efeitos dos fármacos , Músculos/efeitos dos fármacos , Músculos/fisiologia , Neurônios , Técnicas de Patch-Clamp , Pirimidinas/farmacologia , Ratos , Ratos Wistar , Receptores da Neurocinina-1/metabolismo , Centro Respiratório/efeitos dos fármacos , Riluzol/farmacologia , Sono , Veratridina/farmacologia , Vigília
11.
Curr Opin Pulm Med ; 20(6): 527-32, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25221856

RESUMO

PURPOSE OF REVIEW: Our understanding of rapid eye movement (REM) sleep and how it is generated remains a topic of debate. Understanding REM sleep mechanisms is important because several sleep disorders result from disturbances in the neural circuits that control REM sleep and its characteristics. This review highlights recent work concerning how the central nervous system regulates REM sleep, and how the make up and breakdown of these REM sleep-generating circuits contribute to narcolepsy, REM sleep behaviour disorder and sleep apnea. RECENT FINDINGS: A complex interaction between brainstem REM sleep core circuits and forebrain and hypothalamic structures is necessary to generate REM sleep. Cholinergic activation and GABAergic inhibition trigger the activation of subcoeruleus neurons, which form the core of the REM sleep circuit. SUMMARY: Untimely activation of REM sleep circuits leads to cataplexy - involuntary muscle weakness or paralysis - a major symptom of narcolepsy. Degeneration of the REM circuit is associated with excessive muscle activation in REM sleep behaviour disorder. Inappropriate arousal from sleep during obstructive sleep apnea repeatedly disturbs the activity of sleep circuits, particularly the REM sleep circuit.


Assuntos
Estimulantes do Sistema Nervoso Central/uso terapêutico , Hipotálamo/fisiopatologia , Narcolepsia/fisiopatologia , Transtorno do Comportamento do Sono REM/fisiopatologia , Síndromes da Apneia do Sono/fisiopatologia , Sono REM , Neurônios Colinérgicos/efeitos dos fármacos , Neurônios GABAérgicos/efeitos dos fármacos , Humanos , Narcolepsia/etiologia , Vias Neurais/efeitos dos fármacos , Transtorno do Comportamento do Sono REM/complicações , Síndromes da Apneia do Sono/etiologia , Transmissão Sináptica/efeitos dos fármacos
12.
Am J Respir Crit Care Med ; 187(3): 311-9, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23220910

RESUMO

RATIONALE: Inhibition of pharyngeal motoneurons accompanies REM sleep and is a cause of hypoventilation and obstructive sleep apnea in humans. One explanation posits that the neurotransmitters glycine and γ-aminobutyric acid are responsible for REM sleep motor inhibition. However, blockade of that mechanism at cranial motor nuclei increases motor activity in all sleep-wake states, and least of all in REM sleep, arguing against it as a major mechanism of REM sleep pharyngeal motor inhibition. OBJECTIVES: To identify the mechanism of REM sleep inhibition at the hypoglossal motor pool. METHODS: Genioglossus and diaphragm activities were recorded in 34 rats across sleep-wake states. Microdialysis probes were implanted into the hypoglossal motor pool. MEASUREMENTS AND MAIN RESULTS: Here we show that muscarinic receptor antagonism at the hypoglossal motor pool prevents the inhibition of genioglossus activity throughout REM sleep; likewise, with G-protein-coupled inwardly rectifying potassium (GIRK) channel blockade. Importantly, the genioglossus activating effects of these interventions were largest in REM sleep and minimal or often absent in other sleep-wake states. Finally, we showed that muscarinic inhibition of the genioglossus is functionally linked to GIRK channel activation. CONCLUSIONS: We identify a powerful cholinergic-GIRK channel mechanism operating at the hypoglossal motor pool that has its largest inhibitory influence in REM sleep and minimal or no effects in other sleep-wake states. This mechanism is the major cause of REM sleep inhibition at a pharyngeal motor pool critical for effective breathing.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Músculos Faríngeos/fisiologia , Sono REM/fisiologia , Análise de Variância , Animais , Modelos Animais de Doenças , Eletroencefalografia/métodos , Eletromiografia/métodos , Nervo Hipoglosso/fisiologia , Masculino , Músculos Faríngeos/inervação , Faringe/fisiologia , Ratos , Ratos Wistar , Síndromes da Apneia do Sono/fisiopatologia
13.
J Neurosci ; 32(5): 1622-33, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22302804

RESUMO

Serotonin type 1A (5-HT(1A)) receptor-responsive neurons in the pedunculopontine tegmental nucleus (PPTn) become maximally active immediately before and during rapid eye movement (REM) sleep. A prevailing model of REM sleep generation indicates that activation of such neurons contributes significantly to the generation of REM sleep, and if correct then inactivation of such neurons ought to suppress REM sleep. We test this hypothesis using bilateral microperfusion of the 5-HT(1A) receptor agonist 8-hydroxy-2-(di-n-propylamino)tetralin (8-OH-DPAT, 10 µm) into the PPTn; this tool has been shown to selectively silence REM sleep-active PPTn neurons while the activity of wake/REM sleep-active PPTn neurons is unaffected. Contrary to the prevailing model, bilateral microperfusion of 8-OH-DPAT into the PPTn (n = 23 rats) significantly increased REM sleep both as a percentage of the total recording time and sleep time, compared with both within-animal vehicle controls and between-animal time-controls. This increased REM sleep resulted from an increased frequency of REM sleep bouts but not their duration, indicating an effect on mechanisms of REM sleep initiation but not maintenance. Furthermore, an increased proportion of the REM sleep bouts stemmed from periods of low REM sleep drive quantified electrographically. Targeted suppression of 5-HT(1A) receptor-responsive PPTn neurons also increased respiratory rate and respiratory-related genioglossus activity, and increased the frequency and amplitude of the sporadic genioglossus activations occurring during REM sleep. These data indicate that 5-HT(1A) receptor-responsive PPTn neurons normally function to restrain REM sleep by elevating the drive threshold for REM sleep induction, and restrain the expression of respiratory rate and motor activities.


Assuntos
Regulação para Baixo/fisiologia , Atividade Motora/fisiologia , Núcleo Tegmental Pedunculopontino/fisiologia , Receptor 5-HT1A de Serotonina/fisiologia , Mecânica Respiratória/fisiologia , Sono REM/fisiologia , Animais , Eletroencefalografia/métodos , Eletromiografia/métodos , Masculino , Neurônios Motores/metabolismo , Neurônios Motores/fisiologia , Núcleo Tegmental Pedunculopontino/metabolismo , Ratos , Ratos Wistar , Tegmento Mesencefálico/metabolismo , Tegmento Mesencefálico/fisiologia
14.
Expert Opin Ther Targets ; 27(7): 609-626, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37494064

RESUMO

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.


Assuntos
Apneia Obstrutiva do Sono , Humanos , Apneia Obstrutiva do Sono/tratamento farmacológico , Sono/fisiologia , Faringe
15.
Sci Rep ; 13(1): 12299, 2023 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-37516800

RESUMO

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.


Assuntos
Diálise Renal , Apneia Obstrutiva do Sono , Humanos , Animais , Camundongos , Neurônios Motores , Sono , Cafeína , Colina O-Acetiltransferase , Niacinamida , Atividade Motora
16.
Elife ; 122023 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-37458576

RESUMO

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.


Assuntos
Receptores da Neurocinina-1 , Substância P , Camundongos , Animais , Receptores da Neurocinina-1/genética , Neurônios/fisiologia , Bulbo/fisiologia , Respiração , Centro Respiratório/fisiologia , Mamíferos
17.
J Neurosci ; 31(4): 1292-301, 2011 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-21273414

RESUMO

The analgesic properties of the opium poppy Papever somniferum were first mentioned by Hippocrates around 400 BC, and opioid analgesics remain the mainstay of pain management today. These drugs can cause the serious side-effect of respiratory depression that can be lethal with overdose, however the critical brain sites and neurochemical identity of the neurons mediating this depression are unknown. By locally manipulating neurotransmission in the adult rat, we identify the critical site of the medulla, the preBötzinger complex, that mediates opioid-induced respiratory depression in vivo. Here we show that opioids at the preBötzinger complex cause respiratory depression or fatal apnea, with anesthesia and deep-sleep being particularly vulnerable states for opioid-induced respiratory depression. Importantly, we establish that the preBötzinger complex is fully responsible for respiratory rate suppression following systemic administration of opioid analgesics. The site in the medulla most sensitive to opioids corresponds to a region expressing neurokinin-1 receptors, and we show in rhythmically active brainstem section in vitro that neurokinin-1 receptor-expressing preBötzinger complex neurons are selectively inhibited by opioids. In summary, neurokinin-1 receptor-expressing preBötzinger complex neurons constitute the critical site mediating opioid-induced respiratory rate depression, and the key therapeutic target for its prevention or reversal.


Assuntos
Analgésicos Opioides/efeitos adversos , Bulbo/fisiologia , Neurônios/fisiologia , Receptores da Neurocinina-1/biossíntese , Respiração/efeitos dos fármacos , Anestesia , Animais , Apneia/induzido quimicamente , Tronco Encefálico/efeitos dos fármacos , Tronco Encefálico/metabolismo , Depressão Química , Técnicas In Vitro , Masculino , Periodicidade , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/agonistas , Taxa Respiratória/efeitos dos fármacos , Sono , Transmissão Sináptica
18.
J Neurophysiol ; 107(5): 1489-99, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22131386

RESUMO

Nitric oxide (NO) is an important signaling molecule that regulates numerous physiological processes, including activity of respiratory motoneurons. However, molecular mechanism(s) underlying NO modulation of motoneurons remain obscure. Here, we used a combination of in vivo and in vitro recording techniques to examine NO modulation of motoneurons in the hypoglossal motor nucleus (HMN). Microperfusion of diethylamine (DEA; an NO donor) into the HMN of anesthetized adult rats increased genioglossus muscle activity. In the brain slice, whole cell current-clamp recordings from hypoglossal motoneurons showed that exposure to DEA depolarized membrane potential and increased responsiveness to depolarizing current injections. Under voltage-clamp conditions, we found that NO inhibited a Ba(2+)-sensitive background K(+) conductance and activated a Cs(+)-sensitive hyperpolarization-activated inward current (I(h)). When I(h) was blocked with Cs(+) or ZD-7288, the NO-sensitive K(+) conductance exhibited properties similar to TWIK-related acid-sensitive K(+) (TASK) channels, i.e., voltage independent, resistant to tetraethylammonium and 4-aminopyridine but inhibited by methanandamide. The soluble guanylyl cyclase blocker 1H-(1,2,4)oxadiazole(4,3-a)quinoxaline-1-one (ODQ) and the PKG blocker KT-5823 both decreased NO modulation of this TASK-like conductance. To characterize modulation of I(h) in relative isolation, we tested effects of NO in the presence of Ba(2+) to block TASK channels. Under these conditions, NO activated both the instantaneous (I(inst)) and time-dependent (I(ss)) components of I(h). Interestingly, at more hyperpolarized potentials NO preferentially increased I(inst). The effects of NO on I(h) were retained in the presence of ODQ and blocked by the cysteine-specific oxidant N-ethylmaleimide. These results suggest that NO activates hypoglossal motoneurons by cGMP-dependent inhibition of a TASK-like current and S-nitrosylation-dependent activation of I(h).


Assuntos
GMP Cíclico/fisiologia , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Nervo Hipoglosso/fisiologia , Neurônios Motores/fisiologia , Óxido Nítrico/fisiologia , Canais de Potássio de Domínios Poros em Tandem/antagonistas & inibidores , Canais de Potássio de Domínios Poros em Tandem/fisiologia , Animais , Masculino , Proteínas do Tecido Nervoso , Ratos , Ratos Wistar
19.
J Appl Physiol (1985) ; 133(5): 1067-1080, 2022 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-36135952

RESUMO

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.


Assuntos
Medicamentos para o Sistema Respiratório , Apneia Obstrutiva do Sono , Trazodona , Masculino , Feminino , Ratos , Animais , Hormônio Liberador de Tireotropina/farmacologia , Hormônio Liberador de Tireotropina/uso terapêutico , Trazodona/farmacologia , Trazodona/uso terapêutico , Medicamentos para o Sistema Respiratório/farmacologia , Medicamentos para o Sistema Respiratório/uso terapêutico , Nível de Alerta , Sono/fisiologia
20.
Sci Rep ; 12(1): 14896, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-36050440

RESUMO

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.


Assuntos
Nervo Hipoglosso , Apneia Obstrutiva do Sono , Acetilcolina/farmacologia , Animais , Atropina/farmacologia , Feminino , Nervo Hipoglosso/fisiologia , Masculino , Muscarina/farmacologia , Antagonistas Muscarínicos/farmacologia , Fisostigmina/farmacologia , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA