Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Sleep ; 46(9)2023 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-37155728

RESUMO

Long-term use of sodium oxybate (SXB), (also called gamma-hydroxybutyrate [GHB]) attenuates the cataplexy and sleepiness of human narcolepsy. We had previously found that chronic opiate usage in humans and long-term opiate administration to mice significantly increased the number of detected hypocretin/orexin (Hcrt) neurons, decreased their size, and increased Hcrt level in the hypothalamus. We also found that opiates significantly decreased cataplexy in human narcoleptics as well as in narcoleptic mice and that cessation of locus coeruleus neuronal activity preceded and was tightly linked to cataplectic attacks in narcoleptic dogs. We tested the hypothesis that SXB produces changes similar to opiates and now report that chronic SXB administration significantly increased the size of Hcrt neurons, the reverse of what we had seen with opiates in humans and mice. Levels of Hcrt in the hypothalamus were nonsignificantly lower, in contrast to the significant increase in hypothalamic Hcrt level after opiates. SXB decreased tyrosine hydroxylase levels in the locus coeruleus, the major descending projection of the hypocretin system, also the reverse of what we saw with opioids. Therefore despite some similar effects on narcoleptic symptomatology, SXB does not produce anatomical changes similar to those elicited by opiates. Analysis of changes in other links in the cataplexy pathway might further illuminate SXB's mechanism of action on narcolepsy.


Assuntos
Cataplexia , Narcolepsia , Alcaloides Opiáceos , Oxibato de Sódio , Humanos , Camundongos , Animais , Cães , Orexinas/metabolismo , Oxibato de Sódio/farmacologia , Cataplexia/tratamento farmacológico , Cataplexia/metabolismo , Locus Cerúleo/metabolismo , Narcolepsia/tratamento farmacológico , Narcolepsia/metabolismo , Neurônios/metabolismo , Alcaloides Opiáceos/metabolismo
2.
Mol Brain ; 15(1): 21, 2022 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-35246205

RESUMO

It is unclear why orexin-deficient animals, but not wild-type mice, show cataplexy. The current hypothesis predicts simultaneous excitation of cataplexy-inhibiting orexin neurons and cataplexy-inducing amygdala neurons. To test this hypothesis, we measured the activity of putative orexin neurons in orexin-knockout mice during cataplexy episodes using fiber photometry. We created two animal models of orexin-knockout mice with a GCaMP6 fluorescent indicator expressed in putative orexin neurons. We first prepared orexin-knockout mice crossed with transgenic mice carrying a tetracycline-controlled transactivator transgene under the control of the orexin promoter. TetO-GCaMP6 was then introduced into mice via an adeno-associated virus injection or natural crossing. The resulting two models showed restricted expression of GCaMP6 in the hypothalamus, where orexin neurons should be located, and showed excitation to an intruder stress that was similar to that observed in orexin-intact mice in our previous study. The activity of these putative orexin neurons increased immediately before the onset of cataplexy-like behavior but decreased (approximately - 20% of the baseline) during the cataplexy-like episode. We propose that the activity of orexin neurons during cataplexy is moderately inhibited by an unknown mechanism. The absence of cataplexy in wild-type mice may be explained by basal or residual activity-induced orexin release, and emotional stimulus-induced counter activation of orexin neurons may not be necessary. This study will serve as a basis for better treatment of cataplexy in narcolepsy patients.


Assuntos
Cataplexia , Narcolepsia , Animais , Cataplexia/metabolismo , Cataplexia/terapia , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Narcolepsia/metabolismo , Narcolepsia/terapia , Neurônios/metabolismo , Orexinas/metabolismo
3.
Sci Rep ; 10(1): 11024, 2020 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-32620831

RESUMO

This case-control study compared the body composition, resting metabolic rate (RMR), and respiratory quotient (RQ) of narcolepsy patients with those of body mass index (BMI)- gender and age-matched controls. This study included 14 male patients with narcolepsy and 14 matched controls. The narcolepsy patients were subdivided into two subgroups (n = 7/each): those with cataplexy (NT1) and those without cataplexy (NT2). Anthropometric measurements, bioelectric impedance analysis, and indirect calorimetry were used in addition to the calculation of common body-composition indices (conicity index, abdominal volume index, and body adiposity index). Our results showed no significant difference in fat percentage, fat mass, fat-free mass, and TBW among NT1, NT2, and controls (p > 0.05). Compared to matched controls, there was a reduction of muscle mass in both NT1 and NT2 subgroups. The RMR was similar in all groups, while patients in the NT1/NT2 subgroups had a lower RQ, used more fat and fewer carbohydrates during the fasting period. These findings give an insight into the distinctive state of altered metabolism in patients with narcolepsy, especially the resting metabolic rate, which was not altered in NT1 vs. NT2 compared to the controls when matched for BMI, age, and gender.


Assuntos
Cataplexia/metabolismo , Cataplexia/fisiopatologia , Adulto , Metabolismo Basal , Composição Corporal , Calorimetria Indireta , Estudos de Casos e Controles , Impedância Elétrica , Humanos , Masculino , Taxa Respiratória , Adulto Jovem
4.
Elife ; 92020 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-32314734

RESUMO

Orexin/hypocretin-producing and melanin-concentrating hormone-producing (MCH) neurons are co-extensive in the hypothalamus and project throughout the brain to regulate sleep/wakefulness. Ablation of orexin neurons decreases wakefulness and results in a narcolepsy-like phenotype, whereas ablation of MCH neurons increases wakefulness. Since it is unclear how orexin and MCH neurons interact to regulate sleep/wakefulness, we generated transgenic mice in which both orexin and MCH neurons could be ablated. Double-ablated mice exhibited increased wakefulness and decreased both rapid eye movement (REM) and non-REM (NREM) sleep. Double-ablated mice showed severe cataplexy compared with orexin neuron-ablated mice, suggesting that MCH neurons normally suppress cataplexy. Double-ablated mice also showed frequent sleep attacks with elevated spectral power in the delta and theta range, a unique state that we call 'delta-theta sleep'. Together, these results indicate a functional interaction between orexin and MCH neurons in vivo that suggests the synergistic involvement of these neuronal populations in the sleep/wakefulness cycle.


Assuntos
Cataplexia/fisiopatologia , Hormônios Hipotalâmicos , Melaninas , Neurônios/fisiologia , Orexinas , Hormônios Hipofisários , Sono/fisiologia , Vigília/fisiologia , Animais , Cataplexia/metabolismo , Camundongos , Camundongos Transgênicos
5.
Curr Biol ; 29(22): 3803-3813.e5, 2019 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-31679942

RESUMO

Appropriate levels of muscle tone are needed to support waking behaviors such as sitting or standing. However, it is unclear how the brain functions to couple muscle tone with waking behaviors. Cataplexy is a unique experiment of nature in which muscle paralysis involuntarily intrudes into otherwise normal periods of wakefulness. Cataplexy therefore provides the opportunity to identify the circuit mechanisms that couple muscle tone and waking behaviors. Here, we tested the long-standing hypothesis that muscle paralysis during cataplexy is caused by recruitment of the brainstem circuit that induces muscle paralysis during REM sleep. Using behavioral, electrophysiological, and chemogenetic strategies, we found that muscle tone and arousal state can be decoupled by manipulation of the REM sleep circuit (the sublaterodorsal tegmental nucleus [SLD]). First, we show that silencing SLD neurons prevents motor suppression during REM sleep. Second, we show that activating these same neurons promotes cataplexy in narcoleptic (orexin-/-) mice, whereas silencing these neurons prevents cataplexy. Most importantly, we show that SLD neurons can decouple motor activity and arousal state in healthy mice. We show that SLD activation triggers cataplexy-like attacks in wild-type mice that are behaviorally and electrophysiologically indistinguishable from cataplexy in orexin-/- mice. We conclude that the SLD functions to engage arousal-motor synchrony during both wakefulness and REM sleep, and we propose that pathological recruitment of SLD neurons could underlie cataplexy in narcolepsy.


Assuntos
Cataplexia/fisiopatologia , Atividade Motora/fisiologia , Tegmento Mesencefálico/fisiologia , Animais , Nível de Alerta/fisiologia , Encéfalo/fisiologia , Cataplexia/metabolismo , Núcleo Celular , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios Motores/fisiologia , Tono Muscular/fisiologia , Transtorno do Comportamento do Sono REM/fisiopatologia , Sono REM/fisiologia , Tegmento Mesencefálico/metabolismo , Vigília/fisiologia
6.
Neurosci Res ; 142: 16-29, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-29580887

RESUMO

Recent developments in neuroimaging techniques have advanced our understanding of biological mechanisms underpinning narcolepsy. We used MEDLINE to retrieve neuroimaging studies to compare patients with narcolepsy and healthy controls. Thirty-seven studies were identified and demonstrated several replicated abnormalities: (1) gray matter reductions in superior frontal, superior and inferior temporal, and middle occipital gyri, hypothalamus, amygdala, insula, hippocampus, cingulate cortex, thalamus, and nucleus accumbens, (2) decreased fractional anisotropy in white matter of fronto-orbital and cingulate area, (3) reduced brain metabolism or cerebral blood flow in middle and superior frontal, and cingulate cortex (4) increased activity in inferior frontal gyri, insula, amygdala, and nucleus accumbens, and (5) N-acetylaspartate/creatine-phosphocreatine level reduction in hypothalamus. In conclusion, all the replicated findings are still controversial due to the limitations such as heterogeneity or size of the samples and lack of multimodal imaging or follow-up. Thus, future neuroimaging studies should employ multimodal imaging methods in a large sample size of patients with narcolepsy and consider age, duration of disease, age at onset, severity, human leukocyte antigen type, cerebrospinal fluid hypocretin levels, and medication intake in order to elucidate possible neuroimaging characteristic of narcolepsy and identify therapeutic targets.


Assuntos
Encéfalo/metabolismo , Encéfalo/patologia , Cataplexia/metabolismo , Cataplexia/patologia , Narcolepsia/metabolismo , Narcolepsia/patologia , Encéfalo/diagnóstico por imagem , Mapeamento Encefálico , Cataplexia/diagnóstico por imagem , Substância Cinzenta/diagnóstico por imagem , Substância Cinzenta/patologia , Humanos , Narcolepsia/diagnóstico por imagem , Neuroimagem/métodos , Neurônios/metabolismo , Neurônios/patologia
7.
Sleep Med Rev ; 43: 23-36, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30503715

RESUMO

Excessive daytime sleepiness (EDS) and cataplexy are common symptoms of narcolepsy, a sleep disorder associated with the loss of hypocretin/orexin (Hcrt) neurons. Although only a few drugs have received regulatory approval for narcolepsy to date, treatment involves diverse medications that affect multiple biochemical targets and neural circuits. Clinical trials have demonstrated efficacy for the following classes of drugs as narcolepsy treatments: alerting medications (amphetamine, methylphenidate, modafinil/armodafinil, solriamfetol [JZP-110]), antidepressants (tricyclic antidepressants, selective serotonin reuptake inhibitors, serotonin-norepinephrine reuptake inhibitors), sodium oxybate, and the H3-receptor inverse agonist/antagonist pitolisant. Enhanced catecholamine availability and regulation of locus coeruleus (LC) norepinephrine (NE) neuron activity is likely central to the therapeutic activity of most of these compounds. LC NE neurons are integral to sleep/wake regulation and muscle tone; reduced excitatory input to the LC due to compromise of Hcrt/orexin neurons (likely due to autoimmune factors) results in LC NE dysregulation and contributes to narcolepsy/cataplexy symptoms. Agents that increase catecholamines and/or LC activity may mitigate EDS and cataplexy by elevating NE regulation of GABAergic inputs from the amygdala. Consequently, novel medications and treatment strategies aimed at preserving and/or modulating Hcrt/orexin-LC circuit integrity are warranted in narcolepsy/cataplexy.


Assuntos
Cataplexia/tratamento farmacológico , Tratamento Farmacológico/métodos , Imunogenética , Narcolepsia/tratamento farmacológico , Neurobiologia , Adjuvantes Anestésicos/administração & dosagem , Antidepressivos/administração & dosagem , Antidepressivos Tricíclicos/administração & dosagem , Cataplexia/metabolismo , Estimulantes do Sistema Nervoso Central/administração & dosagem , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Modafinila/administração & dosagem , Narcolepsia/metabolismo , Oxibato de Sódio/administração & dosagem
8.
Sleep ; 41(9)2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29868885

RESUMO

Study Objective: Narcolepsy type 1 (NT1) is considered a chronic, incurable disease. Excessive daytime sleepiness (EDS) is typically the most troublesome symptom, and more difficult to control by pharmacologic treatment than cataplexy. Although many NT1 patients are monitored by regular follow-ups, the purported relentless persistence of EDS has rarely been the object of longitudinal studies. Methods: Retrospective analysis of 26 well-defined hypocretin-deficient NT1 patients who underwent longitudinal assessments of Epworth sleepiness scale (ESS) scores under stable pharmacotherapy. We present detailed case reports of four patients with unusual spontaneous improvement. Results: Over a mean observation period of 5 years, changes in ESS scores between first and last examination were ≤4 points in 19 patients (73%). Three patients deteriorated by 5 points, four patients ameliorated by 7-11 points. Among the latter, subjective sleepiness resolved in all four patients, and three of them continued showing ESS scores <11 after cessation of their pharmacotherapy. Without therapy, two patients did not fulfill anymore the ICSD-3 multiple sleep latency test criteria (mean sleep latency >8 minutes), one of whom did not fall asleep during maintenance of wakefulness test. Multiple linear regression analysis identified higher cerebrospinal fluid (CSF) hypocretin level (p < 0.001) and absence of fragmented nighttime sleep (p = 0.001) as independent associates of EDS improvement. Conclusions: The longitudinal course of NT1-related sleepiness is not invariably stable, but included spontaneous deterioration or improvement in 27%. Spontaneous improvement can persist after treatment discontinuation and resemble remission. Milder hypocretin deficiency and good nighttime sleep may predict a more favorable disease course.


Assuntos
Narcolepsia/genética , Narcolepsia/metabolismo , Orexinas/deficiência , Orexinas/genética , Remissão Espontânea , Latência do Sono/fisiologia , Adulto , Cataplexia/genética , Cataplexia/metabolismo , Cataplexia/fisiopatologia , Distúrbios do Sono por Sonolência Excessiva/genética , Distúrbios do Sono por Sonolência Excessiva/metabolismo , Distúrbios do Sono por Sonolência Excessiva/fisiopatologia , Feminino , Humanos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Narcolepsia/fisiopatologia , Polissonografia/tendências , Estudos Retrospectivos , Sono/fisiologia , Vigília/fisiologia
9.
Proc Natl Acad Sci U S A ; 115(23): 6046-6051, 2018 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-29784823

RESUMO

Narcolepsy-cataplexy is a chronic neurological disorder caused by loss of orexin (hypocretin)-producing neurons, associated with excessive daytime sleepiness, sleep attacks, cataplexy, sleep paralysis, hypnagogic hallucinations, and fragmentation of nighttime sleep. Currently, human narcolepsy is treated by providing symptomatic therapies, which can be associated with an array of side effects. Although peripherally administered orexin does not efficiently penetrate the blood-brain barrier, centrally delivered orexin can effectively alleviate narcoleptic symptoms in animal models. Chronic intrathecal drug infusion through an implantable pump is a clinically available strategy to treat a number of neurological diseases. Here we demonstrate that the narcoleptic symptoms of orexin knockout mice can be reversed by lumbar-level intrathecal orexin delivery. Orexin was delivered via a chronically implanted intrathecal catheter at the upper lumbar level. The computed tomographic scan confirmed that intrathecally administered contrast agent rapidly moved from the spinal cord to the brain. Intrathecally delivered orexin was detected in the brain by radioimmunoassay at levels comparable to endogenous orexin levels. Cataplexy and sleep-onset REM sleep were significantly decreased in orexin knockout mice during and long after slow infusion of orexin (1 nmol/1 µL/h). Sleep/wake states remained unchanged both quantitatively as well as qualitatively. Intrathecal orexin failed to induce any changes in double orexin receptor-1 and -2 knockout mice. This study supports the concept of intrathecal orexin delivery as a potential therapy for narcolepsy-cataplexy to improve the well-being of patients.


Assuntos
Narcolepsia/tratamento farmacológico , Orexinas/administração & dosagem , Orexinas/farmacologia , Animais , Encéfalo/fisiologia , Cataplexia/tratamento farmacológico , Cataplexia/metabolismo , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Orexinas/metabolismo , Sono/efeitos dos fármacos , Transtornos do Sono do Ritmo Circadiano/tratamento farmacológico , Transtornos do Sono do Ritmo Circadiano/metabolismo , Vigília/efeitos dos fármacos
10.
J Neurosci ; 37(15): 3995-4006, 2017 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-28235898

RESUMO

Narcolepsy is characterized by chronic sleepiness and cataplexy-sudden muscle paralysis triggered by strong, positive emotions. This condition is caused by a lack of orexin (hypocretin) signaling, but little is known about the neural mechanisms that mediate cataplexy. The amygdala regulates responses to rewarding stimuli and contains neurons active during cataplexy. In addition, lesions of the amygdala reduce cataplexy. Because GABAergic neurons of the central nucleus of the amygdala (CeA) target brainstem regions known to regulate muscle tone, we hypothesized that these cells promote emotion-triggered cataplexy. We injected adeno-associated viral vectors coding for Cre-dependent DREADDs or a control vector into the CeA of orexin knock-out mice crossed with vGAT-Cre mice, resulting in selective expression of the excitatory hM3 receptor or the inhibitory hM4 receptor in GABAergic neurons of the CeA. We measured sleep/wake behavior and cataplexy after injection of saline or the hM3/hM4 ligand clozapine-N-oxide (CNO) under baseline conditions and under conditions that should elicit positive emotions. In mice expressing hM3, CNO approximately doubled the amount of cataplexy in the first 3 h after dosing under baseline conditions. Rewarding stimuli (chocolate or running wheels) also increased cataplexy, but CNO produced no further increase. In mice expressing hM4, CNO reduced cataplexy in the presence of chocolate or running wheels. These results demonstrate that GABAergic neurons of the CeA are sufficient and necessary for the production of cataplexy in mice, and they likely are a key part of the mechanism through which positive emotions trigger cataplexy.SIGNIFICANCE STATEMENT Cataplexy is one of the major symptoms of narcolepsy, but little is known about how strong, positive emotions trigger these episodes of muscle paralysis. Prior research shows that amygdala neurons are active during cataplexy and cataplexy is reduced by lesions of the amygdala. We found that cataplexy is substantially increased by selective activation of GABAergic neurons in the central nucleus of the amygdala (CeA). We also demonstrate that inhibition of these neurons reduces reward-promoted cataplexy. These results build upon prior work to establish the CeA as a crucial element in the neural mechanisms of cataplexy. These results demonstrate the importance of the CeA in regulating responses to rewarding stimuli, shedding light on the broader neurobiology of emotions and motor control.


Assuntos
Cataplexia/genética , Cataplexia/metabolismo , Núcleo Central da Amígdala/metabolismo , Neurônios GABAérgicos/metabolismo , Animais , Locomoção/fisiologia , Masculino , Camundongos , Camundongos Knockout
11.
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
12.
Eur J Neurosci ; 43(5): 681-8, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26741960

RESUMO

Narcolepsy is a chronic sleep disorder linked to the loss of orexin-producing neurons in the hypothalamus. Cataplexy, a sudden loss of muscle tone during waking, is an important distinguishing symptom of narcolepsy and it is often triggered by strong emotions. The neural circuit underlying cataplexy attacks is not known, but is likely to involve the amygdala, a region implicated in regulating emotions. In mice models of narcolepsy, transfer of the orexin gene into surrogate neurons has been successful in ameliorating narcoleptic symptoms. However, it is not known whether this method also blocks cataplexy triggered by strong emotions. To examine this possibility, the gene encoding mouse prepro-orexin was transferred into amygdala neurons of orexin-knockout (KO) mice (rAAV-orexin; n = 8). Orexin-KO mice that did not receive gene transfer (no-rAAV; n = 7) or received only the reporter gene (rAAV-GFP; n = 7) served as controls. Three weeks later, the animal's sleep and behaviour were recorded at night (no-odour control night), followed by another recording at night in the presence of predator odour (odour night). Orexin-KO mice given the orexin gene transfer into surrogate amygdala neurons had significantly less spontaneous bouts of cataplexy, and predator odour did not induce cataplexy compared with control mice. Moreover, the mice with orexin gene transfer were awake more during the odour night. These results demonstrate that orexin gene transfer into amygdala neurons can suppress both spontaneous and emotion-induced cataplexy attacks in narcoleptic mice. It suggests that manipulating amygdala pathways is a potential strategy for treating cataplexy in narcolepsy.


Assuntos
Tonsila do Cerebelo/metabolismo , Cataplexia/metabolismo , Orexinas/metabolismo , Tonsila do Cerebelo/fisiologia , Animais , Cataplexia/terapia , Emoções , Feminino , Terapia Genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Orexinas/genética , Sono REM
13.
PLoS One ; 10(11): e0142432, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26599765

RESUMO

The proper functioning of the mesolimbic reward system is largely dependent on the neurotransmitter dopamine. Recent evidence suggests that the hypocretin system has significant projections to this reward system. We examined the distinct effects of reduced dopamine or reduced hypocretin levels on reward activity in patients with Parkinson's disease, dopamine deficient, as well as patients with narcolepsy-cataplexy, hypocretin depleted, and healthy controls. Participants performed a simple game-like task while high-density electroencephalography was recorded. Topography and timing of event-related potentials for both reward cue, and reward feedback was examined across the entire dataset. While response to reward cue was similar in all groups, two distinct time points were found to distinguish patients and controls for reward feedback. Around 160 ms both patient groups had reduced ERP amplitude compared to controls. Later at 250 ms, both patient groups also showed a clear event-related potential (ERP), which was absent in controls. The initial differences show that both patient groups show a similar, blunted response to reward delivery. The second potential corresponds to the classic feedback-related negativity (FRN) potential which relies on dopamine activity and reflects reward prediction-error signaling. In particular the mismatch between predicted reward and reward subsequently received was significantly higher in PD compared to NC, independent of reward magnitude and valence. The intermediate FRN response in NC highlights the contribution of hypocretin in reward processing, yet also shows that this is not as detrimental to the reward system as in Parkinson's. Furthermore, the inability to generate accurate predictions in NC may explain why hypocretin deficiency mediates cataplexy triggered by both positive and negative emotions.


Assuntos
Cataplexia/fisiopatologia , Dopamina/fisiologia , Eletroencefalografia , Potenciais Evocados P300 , Narcolepsia/fisiopatologia , Orexinas/fisiologia , Doença de Parkinson/fisiopatologia , Recompensa , Adulto , Idoso , Comportamento , Estudos de Casos e Controles , Cataplexia/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Narcolepsia/metabolismo , Orexinas/deficiência , Doença de Parkinson/metabolismo , Reprodutibilidade dos Testes , Processamento de Sinais Assistido por Computador
14.
PLoS One ; 10(10): e0140520, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26474479

RESUMO

Narcolepsy type 1 is associated with loss of orexin neurons, sleep-wake derangements, cataplexy, and a wide spectrum of alterations in other physiological functions, including energy balance, cardiovascular, and respiratory control. It is unclear which narcolepsy signs are directly related to the lack of orexin neurons or are instead modulated by dysfunction of other neurotransmitter systems physiologically controlled by orexin neurons, such as the histamine system. To address this question, we tested whether some of narcolepsy signs would be detected in mice lacking histamine signaling (HDC-KO). Moreover, we studied double-mutant mice lacking both histamine signaling and orexin neurons (DM) to evaluate whether the absence of histamine signaling would modulate narcolepsy symptoms produced by orexin deficiency. Mice were instrumented with electrodes for recording the electroencephalogram and electromyogram and a telemetric arterial pressure transducer. Sleep attacks fragmenting wakefulness, cataplexy, excess rapid-eye-movement sleep (R) during the activity period, and enhanced increase of arterial pressure during R, which are hallmarks of narcolepsy in mice, did not occur in HDC-KO, whereas they were observed in DM mice. Thus, these narcolepsy signs are neither caused nor abrogated by the absence of histamine. Conversely, the lack of histamine produced obesity in HDC-KO and to a greater extent also in DM. Moreover, the regularity of breath duration during R was significantly increased in either HDC-KO or DM relative to that in congenic wild-type mice. Defects of histamine transmission may thus modulate the metabolic and respiratory phenotype of murine narcolepsy.


Assuntos
Histamina/metabolismo , Narcolepsia/metabolismo , Narcolepsia/patologia , Neurônios/metabolismo , Orexinas/deficiência , Fenótipo , Transdução de Sinais , Animais , Peso Corporal , Sistema Cardiovascular/fisiopatologia , Cataplexia/metabolismo , Cataplexia/patologia , Cataplexia/fisiopatologia , Ingestão de Energia , Feminino , Técnicas de Inativação de Genes , Histidina Descarboxilase/deficiência , Histidina Descarboxilase/genética , Masculino , Camundongos , Respiração , Sono , Vigília
15.
J Psychopharmacol ; 29(6): 744-9, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25735989

RESUMO

The sleep disorder narcolepsy is caused by the loss of orexinergic neurones in the lateral hypothalamus. A troublesome symptom of narcolepsy is cataplexy, the sudden loss of muscle tone in response to strong emotions. It can be alleviated by antidepressants and sodium oxybate (γ-hydroxybutyric acid (GHB)). It is likely that the noradrenergic nucleus locus coeruleus (LC) is involved since it is essential for the maintenance of muscle tone, and ceases to fire during cataplectic attacks. Furthermore, alpha-2 adrenoceptors proliferate in the LC in cataplexy, probably due to 'heterologous denervation supersensitivity' resulting from the loss/weakening of the orexinergic input to the LC. This would lead to the sensitization of the autoinhibition mechanism of LC neurones mediated by inhibitory alpha-2 adrenoceptors ('autoreceptors'). Thus the excitatory input from the amygdala to the LC, activated by an emotional stimulus, would lead to the 'switching off' of LC activity via the supersensitive auto-inhibition mechanism. GHB is an agonist at both γ-aminobutyric acid (GABA) GABA (B) and GHB receptors that may be a subtype of an extrasynaptic GABA(A) receptor. GHB may prevent a cataplectic attack by dampening the tone of LC neurones via the stimulation of inhibitory extrasynaptic GABA receptors in the LC, and thus increasing the threshold for autoinhibition.


Assuntos
Cataplexia/tratamento farmacológico , Hidroxibutiratos/farmacologia , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Cataplexia/metabolismo , Emoções/efeitos dos fármacos , Humanos , Locus Cerúleo/efeitos dos fármacos , Locus Cerúleo/metabolismo , Músculos/efeitos dos fármacos , Músculos/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Receptores de GABA-B/metabolismo , Sono/efeitos dos fármacos , Oxibato de Sódio/farmacologia , Ácido gama-Aminobutírico/metabolismo
16.
CNS Spectr ; 19 Suppl 1: 25-33; quiz 25-7, 34, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25403789

RESUMO

The medications used to treat narcolepsy are targeted toward alleviating symptoms such as excessive sleepiness and cataplexy. The cause of this neurological sleep disorder is still not completely clear, though a destruction of hypocretin/orexin neurons has been implicated. The destruction of these neurons is linked to inactivity of neurotransmitters including histamine, norepinephrine, acetylcholine, and serotonin, causing a disturbance in the sleep/wake cycles of narcoleptic patients. Stimulants and MAOIs have traditionally been used to counteract excessive daytime sleepiness and sleep attacks by inhibiting the breakdown of catecholamines. Newer drugs, called wake-promoting agents, have recently become first-line agents due to their better side-effect profile, efficacy, and lesser potential for abuse. These agents similarly inhibit reuptake of dopamine, but have a novel mechanism of action, as they have been found to increase neuronal activity in the tuberomamillary nucleus and in orexin neurons. Sodium oxybate, a sodium salt of gamma-hydroxybutyrate (GHB), is another class that is used to treat many symptoms of narcolepsy, and is the only U.S. Food and Drug Administration (FDA)-approved medication for cataplexy. It has a different mechanism of action than either stimulants or wake-promoting agents, as it binds to its own unique receptor. Antidepressants, like selective serotonin re-uptake inhibitors (SSRIs) and tricyclic antidepressants (TCAs), have also been used, as similar to stimulants, they inhibit reuptake of specific catecholamines. In this article, we seek to review the mechanisms behind these classes of drugs in relation to the proposed pathophysiology of narcolepsy. Appropriate clinical strategies will be discussed, including specific combinations of medications that have been shown to be effective.


Assuntos
Antidepressivos Tricíclicos/uso terapêutico , Estimulantes do Sistema Nervoso Central/uso terapêutico , Inibidores da Captação de Dopamina/uso terapêutico , Inibidores da Monoaminoxidase/uso terapêutico , Narcolepsia/tratamento farmacológico , Promotores da Vigília/uso terapêutico , Antidepressivos/uso terapêutico , Cataplexia/tratamento farmacológico , Cataplexia/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Narcolepsia/metabolismo , Neuropeptídeos/metabolismo , Orexinas , Oxibato de Sódio/uso terapêutico
17.
J Sleep Res ; 23(4): 425-31, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24650212

RESUMO

Narcolepsy with cataplexy is caused by a deficiency in the production of hypocretin/orexin, which regulates sleep and wakefulness, and also influences appetite, neuroendocrine functions and metabolism. In this case-control study, 11 patients with narcolepsy with cataplexy and 11 healthy adults underwent an oral glucose tolerance test, and dexamethasone suppression/corticotropin-releasing hormone stimulation test. The average age of patients and controls was 35.1 ± 13.2 and 41.0 ± 2.9 years, respectively, body mass index was 28.1 ± 6.6 and 25.5 ± 4.7 kg m(-2) . We did not find evidence of a significantly increased prevalence of disturbed glucose tolerance in patients with narcolepsy. After hypothalamo-pituitary-adrenal axis suppression, the number of non-suppressors did not differ between the groups, indicating normal negative feedback sensitivity. The level of cortisol after dexamethasone suppression was significantly lower in patients with narcolepsy, suggesting a slight basal downregulation and/or a slightly increased negative feedback sensitivity of the major endocrine stress system in narcolepsy. Following corticotropin-releasing hormone stimulation, there were no significant differences in levels of adrenocorticotropic hormone or cortisol, and in adrenocortical responsivity to adrenocorticotropic hormone. Finally, patients with narcolepsy displayed significantly higher plasma levels of tumour necrosis factor alpha, soluble tumour necrosis factor receptor p55, soluble tumour necrosis factor receptor p75 and interleukin 6 after adjustment for body mass index. The present study confirms that narcolepsy by itself is not associated with disturbances of glucose metabolism, but goes along with a subtle dysregulation of inflammatory cytokine production. We also found that dynamic hypothalamo-pituitary-adrenal system response is not altered, whereas negative feedback to dexamethasone might be slightly enhanced.


Assuntos
Glicemia/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Narcolepsia/sangue , Narcolepsia/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Fator de Necrose Tumoral alfa/sangue , Hormônio Adrenocorticotrópico/sangue , Adulto , Índice de Massa Corporal , Estudos de Casos e Controles , Cataplexia/sangue , Cataplexia/complicações , Cataplexia/metabolismo , Hormônio Liberador da Corticotropina/farmacologia , Dexametasona/farmacologia , Feminino , Teste de Tolerância a Glucose , Humanos , Hidrocortisona/sangue , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Interleucina-6/metabolismo , Masculino , Narcolepsia/complicações , Sistema Hipófise-Suprarrenal/efeitos dos fármacos
18.
Brain Behav Immun ; 37: 260-6, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24394344

RESUMO

Recent advances in the identification of susceptibility genes and environmental exposures provide strong support that narcolepsy-cataplexy is an immune-mediated disease. Only few serum cytokine studies with controversial results were performed in narcolepsy and none in the cerebrospinal fluid. We measured a panel of 12 cytokines by a proteomic approach in the serum of 35 patients with narcolepsy-cataplexy compared to 156 healthy controls, and in the cerebrospinal fluid of 34 patients with narcolepsy-cataplexy compared to 17 non-narcoleptic patients; and analyzed the effect of age, duration and severity of disease on the cytokine levels. After multiple adjustments we reported lower serum IL-2, IL-8, TNF-α, MCP-1 and EGF levels, and a tendency for higher IL-4 level in narcolepsy compared to controls. Significant differences were only found for IL-4 in cerebrospinal fluid, being higher in narcolepsy. Positive correlations were found in serum between IL-4, daytime sleepiness, and cataplexy frequency. The expression of some pro-inflammatory cytokines (MCP-1, VEGF, EGF, IL2, IL-1ß, IFN-γ) in either serum or CSF was negatively correlated with disease severity and duration. No correlation was found for any specific cytokine in 18 of the patients with narcolepsy with peripheral and central samples collected the same day. Significant decreased pro/anti-inflammatory cytokine profiles were found at peripheral and central levels in narcolepsy, together with a T helper 2/Th1 serum cytokine secretion imbalance. To conclude, we showed some evidence for alterations in the cytokine profile in patients with narcolepsy-cataplexy compared to controls at peripheral and central levels, with the potential role of IL-4 and significant Th1/2 imbalance in the pathophysiology of narcolepsy.


Assuntos
Cataplexia/metabolismo , Citocinas/metabolismo , Narcolepsia/metabolismo , Adolescente , Adulto , Idoso , Estudos de Casos e Controles , Cataplexia/sangue , Cataplexia/líquido cefalorraquidiano , Cataplexia/complicações , Criança , Pré-Escolar , Citocinas/sangue , Citocinas/líquido cefalorraquidiano , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Narcolepsia/sangue , Narcolepsia/líquido cefalorraquidiano , Narcolepsia/complicações , Índice de Gravidade de Doença , Adulto Jovem
19.
J Sleep Res ; 23(1): 98-106, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24033681

RESUMO

Narcolepsy with cataplexy (NC) is a lifelong disorder caused by loss of hypothalamic hypocretin/orexin (HCRT) neurones, often starting in childhood. NC patients show altered control of heart rate (HR) and a normotensive non-dipper blood pressure (BP) profile, but the natural history and prognostic significance of these alterations remain unclear. Similar alterations have been observed in HCRT-ataxin-3 transgenic (TG) NC mice lacking HCRT neurones, but studies have been limited to young adult individuals <4 months of age. Here we evaluated long-term effects of NC on derangements in the wake-sleep state and cardiovascular control by studying middle-aged TG. We chronically instrumented TG and wild-type mice aged 10-11 months with electrodes for sleep scoring and a telemetric transducer for BP and HR measurements. We then recorded mice in freely behaving conditions. TG showed a NC phenotype including fragmentation of wakefulness, reduced latency to rapid eye movement sleep (REMS) and cataplexy-like events. TG also showed blunted BP decline on entering non-rapid eye movement sleep (NREMS), enhanced BP increase on passing to REMS, increased HR, and blunted changes in HR upon arousal and awakening from NREMS. Histological and ultrastructural analysis of cardiovascular and renal tissue did not reveal evidence of subclinical hypertensive organ damage. These data indicate that HCRT neurone loss in TG causes alterations in wake-sleep behaviour and cardiovascular control that are not peculiar to the beginning of the disease but are maintained at least up to middle age. These alterations are similar to those in adult NC patients, but do not produce early subclinical damage to the heart and kidneys.


Assuntos
Envelhecimento/fisiologia , Cataplexia/fisiopatologia , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Neuropeptídeos/deficiência , Sono/fisiologia , Animais , Pressão Sanguínea/fisiologia , Peso Corporal , Cataplexia/metabolismo , Frequência Cardíaca/fisiologia , Masculino , Camundongos , Neurônios/metabolismo , Orexinas , Fenótipo , Sono REM , Vigília/fisiologia
20.
Sleep Med ; 14(12): 1272-6, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24157100

RESUMO

BACKGROUND: Narcolepsy is a neuropsychiatric disorder characterized by excessive daytime sleepiness, cataplexy, hypnagogic hallucinations, and abnormal rapid eye movement (REM) sleep. Tumor necrosis factor α (TNF α) and its cognate receptors have been reported to be involved in the pathophysiology of narcolepsy in addition to the HLA antigen system. Our study aimed to determine if the TNF-α system was associated with narcolepsy in our patients. METHODS: We first measured the plasma level of TNF α in 56 narcoleptic patients and 53 control subjects using a highly sensitive enzyme-linked immunosorbent assay. We next determined the genotype of three single nucleotide polymorphisms (SNPs) (T-1031C, C-863A, and C-857T) at the promoter region of the TNF-α gene and one missense SNP (T587G, M196R) at the exon 6 of the tumor necrosis factor receptor 2 gene, TNFR2, in a sample of 75 narcoleptic patients and 201 control subjects by direct sequencing analysis. RESULTS: We found a significant elevation of plasma level of TNF α in patients with narcolepsy compared with the control subjects (4.64pg/mL vs 1.06pg/mL; P=.0013). However, we did not find significant differences between these two groups in the allelic and genotypic distributions of the investigated polymorphisms. CONCLUSIONS: Our study suggests that an increased TNF-α level was associated with narcolepsy in our patients, and that chronic inflammation due to various factors might have led to the increased TNF-α levels found in our patients.


Assuntos
Narcolepsia/genética , Narcolepsia/metabolismo , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/genética , Adolescente , Adulto , Cataplexia/genética , Cataplexia/metabolismo , Criança , Feminino , Frequência do Gene , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Mutação de Sentido Incorreto/genética , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas/genética , Receptores Tipo II do Fator de Necrose Tumoral/genética , Taiwan , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...