Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Brain ; 145(6): 2018-2030, 2022 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-35552381

RESUMO

Narcolepsy with cataplexy or narcolepsy type 1 is a disabling chronic sleep disorder resulting from the destruction of orexinergic neurons in the hypothalamus. The tight association of narcolepsy with HLA-DQB1*06:02 strongly suggest an autoimmune origin to this disease. Furthermore, converging epidemiological studies have identified an increased incidence for narcolepsy in Europe following Pandemrix® vaccination against the 2009-2010 pandemic 'influenza' virus strain. The potential immunological link between the Pandemrix® vaccination and narcolepsy remains, however, unknown. Deciphering these mechanisms may reveal pathways potentially at play in most cases of narcolepsy. Here, we developed a mouse model allowing to track and study the T-cell response against 'influenza' virus haemagglutinin, which was selectively expressed in the orexinergic neurons as a new self-antigen. Pandemrix® vaccination in this mouse model resulted in hypothalamic inflammation and selective destruction of orexin-producing neurons. Further investigations on the relative contribution of T-cell subsets in this process revealed that haemagglutinin-specific CD4 T cells were necessary for the development of hypothalamic inflammation, but insufficient for killing orexinergic neurons. Conversely, haemagglutinin-specific CD8 T cells could not initiate inflammation but were the effectors of the destruction of orexinergic neurons. Additional studies revealed pathways potentially involved in the disease process. Notably, the interferon-γ pathway was proven essential, as interferon-γ-deficient CD8 T cells were unable to elicit the loss of orexinergic neurons. Our work demonstrates that an immunopathological process mimicking narcolepsy can be elicited by immune cross-reactivity between a vaccine antigen and a neuronal self-antigen. This process relies on a synergy between autoreactive CD4 and CD8 T cells for disease development. This work furthers our understanding of the mechanisms and pathways potentially involved in the development of a neurological side effect due to a vaccine and, likely, to narcolepsy in general.


Assuntos
Autoimunidade , Vacinas contra Influenza , Narcolepsia , Animais , Autoantígenos , Hemaglutininas , Inflamação/complicações , Vacinas contra Influenza/efeitos adversos , Interferon gama , Camundongos , Narcolepsia/induzido quimicamente , Neurônios , Orexinas , Linfócitos T/imunologia , Vacinação/efeitos adversos
2.
Proc Natl Acad Sci U S A ; 113(39): 10956-61, 2016 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-27621438

RESUMO

Narcolepsy with cataplexy is a rare and severe sleep disorder caused by the destruction of orexinergic neurons in the lateral hypothalamus. The genetic and environmental factors associated with narcolepsy, together with serologic data, collectively point to an autoimmune origin. The current animal models of narcolepsy, based on either disruption of the orexinergic neurotransmission or neurons, do not allow study of the potential autoimmune etiology. Here, we sought to generate a mouse model that allows deciphering of the immune mechanisms leading to orexin(+) neuron loss and narcolepsy development. We generated mice expressing the hemagglutinin (HA) as a "neo-self-antigen" specifically in hypothalamic orexin(+) neurons (called Orex-HA), which were transferred with effector neo-self-antigen-specific T cells to assess whether an autoimmune process could be at play in narcolepsy. Given the tight association of narcolepsy with the human leukocyte antigen (HLA) HLA-DQB1*06:02 allele, we first tested the pathogenic contribution of CD4 Th1 cells. Although these T cells readily infiltrated the hypothalamus and triggered local inflammation, they did not elicit the loss of orexin(+) neurons or clinical manifestations of narcolepsy. In contrast, the transfer of cytotoxic CD8 T cells (CTLs) led to both T-cell infiltration and specific destruction of orexin(+) neurons. This phenotype was further aggravated upon repeated injections of CTLs. In situ, CTLs interacted directly with MHC class I-expressing orexin(+) neurons, resulting in cytolytic granule polarization toward neurons. Finally, drastic neuronal loss caused manifestations mimicking human narcolepsy, such as cataplexy and sleep attacks. This work demonstrates the potential role of CTLs as final effectors of the immunopathological process in narcolepsy.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Citotoxicidade Imunológica , Narcolepsia/imunologia , Narcolepsia/patologia , Neurônios/patologia , Orexinas/metabolismo , Animais , Autoanticorpos/metabolismo , Autoantígenos/metabolismo , Comunicação Celular , Hemaglutininas/metabolismo , Hipotálamo/metabolismo , Inflamação/patologia , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Fenótipo , Linfócitos T Citotóxicos/metabolismo , Células Th1/metabolismo
3.
J Neurosci ; 37(9): 2268-2272, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28250055

RESUMO

Early in 1998, we (de Lecea et al., 1998) and others (Sakurai et al., 1998) described the same hypothalamic neuropeptides, respectively called the hypocretins or orexins, which were discovered using two different approaches. In December of that year, we published the subject of this commentary in the Journal of Neuroscience: a highly detailed anatomical description of the extensive axonal projections of the hypocretin/orexin neurons. Although the function of this system was unknown at the time, a large body of literature today attests that the hypocretin/orexin neuropeptides play important roles in multiple physiological functions, particularly in sleep/wake regulation. Neuroanatomical studies are rarely frontline news, but the citation rate of this paper underscores the critical nature of such basic research. Based in part on this detailed description, the hypocretin/orexin neuropeptides have since been studied in many different areas of neuroscience research, including sleep/wake regulation, feeding, addiction, reward and motivation, anxiety and depression, cardiovascular regulation, pain, migraine, and neuroendocrine regulation, including reproduction. Thus, this paper has had a surprisingly broad impact on neuroscience research, particularly since it was originally rejected by the Journal!


Assuntos
Encéfalo/metabolismo , Neurônios/metabolismo , Neurociências/tendências , Orexinas/fisiologia , Publicações Periódicas como Assunto , Animais , História do Século XX , História do Século XXI , Humanos , Neurociências/história , Orexinas/história , Publicações Periódicas como Assunto/história , Publicações Periódicas como Assunto/tendências
4.
J Sleep Res ; 24(3): 309-19, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25524602

RESUMO

Studying paradoxical sleep homeostasis requires the specific and efficient deprivation of paradoxical sleep and the evaluation of the subsequent recovery period. With this aim, the small-platforms-over-water technique has been used extensively in rats, but only rare studies were conducted in mice, with no sleep data reported during deprivation. Mice are used increasingly with the emergence of transgenic mice and technologies such as optogenetics, raising the need for a reliable method to manipulate paradoxical sleep. To fulfil this need, we refined this deprivation method and analysed vigilance states thoroughly during the entire protocol. We also studied activation of hypocretin/orexin and melanin-concentrating hormone neurones using Fos immunohistochemistry to verify whether mechanisms regulating paradoxical sleep in mice are similar to those in rats. We showed that 48 h of deprivation was highly efficient, with a residual amount of paradoxical sleep of only 2.2%. Slow wave sleep and wake quantities were similar to baseline, except during the first 4 h of deprivation, where slow wave sleep was strongly reduced. After deprivation, we observed a 124% increase in paradoxical sleep quantities during the first hour of rebound. In addition, 34% of hypocretin/orexin neurones were activated during deprivation, whereas melanin-concentrated hormone neurones were activated only during paradoxical sleep rebound. Corticosterone level showed a twofold increase after deprivation and returned to baseline level after 4 h of recovery. In summary, a fairly selective deprivation and a significant rebound of paradoxical sleep can be obtained in mice using the small-platforms-over-water method. As in rats, rebound is accompanied by a selective activation of melanin-concentrating hormone neurones.


Assuntos
Hormônios Hipotalâmicos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Melaninas/metabolismo , Neurônios/fisiologia , Neuropeptídeos/metabolismo , Hormônios Hipofisários/metabolismo , Privação do Sono/fisiopatologia , Sono REM/fisiologia , Água , Animais , Atenção/fisiologia , Corticosterona/metabolismo , Homeostase , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Orexinas , Polissonografia , Ratos , Sono/fisiologia , Fatores de Tempo , Vigília/fisiologia
5.
Int J Neuropsychopharmacol ; 17(1): 157-68, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23702225

RESUMO

Insomnia is a serious medical and social problem, its prevalence in the general population ranges from 9 to 35% depending on the country and assessment method. Often, patients are subject to inappropriate and therefore dangerous pharmacotherapies that include prolonged administration of hypnotic drugs, benzodiazepines and other GABAA receptor modulators. This usually does not lead to a satisfactory improvement in patients' clinical states and may cause lifelong drug dependence. Brain state transitions require the coordinated activity of numerous neuronal pathways and brain structures. It is thought that orexin-expressing neurons play a crucial role in this process. Due to their interaction with the sleep-wake-regulating neuronal population, they can activate vigilance-promoting regions and prevent unwanted sleep intrusions. Understanding the multiple orexin modulatory effects is crucial in the context of pathogenesis of insomnia and should lead to the development of novel treatments. An important step in this process was the synthesis of dual antagonists of orexin receptors. Crucially, these drugs, as opposed to benzodiazepines, do not change the sleep architecture and have limited side-effects. This new pharmacological approach might be the most appropriate to treat insomnia.


Assuntos
Acetamidas/uso terapêutico , Azepinas/uso terapêutico , Benzofuranos/uso terapêutico , Isoquinolinas/uso terapêutico , Antagonistas dos Receptores de Orexina , Piperidinas/uso terapêutico , Pirimidinas/uso terapêutico , Distúrbios do Início e da Manutenção do Sono/tratamento farmacológico , Tiazóis/uso terapêutico , Triazóis/uso terapêutico , Acetamidas/farmacocinética , Acetamidas/farmacologia , Animais , Azepinas/farmacologia , Benzofuranos/farmacocinética , Benzofuranos/farmacologia , Humanos , Isoquinolinas/farmacocinética , Isoquinolinas/farmacologia , Modelos Biológicos , Receptores de Orexina/fisiologia , Piperidinas/farmacocinética , Piperidinas/farmacologia , Pirimidinas/farmacocinética , Pirimidinas/farmacologia , Sono/efeitos dos fármacos , Sono/fisiologia , Tiazóis/farmacocinética , Tiazóis/farmacologia , Triazóis/farmacologia , Vigília/efeitos dos fármacos , Vigília/fisiologia
6.
Pflugers Arch ; 463(1): 43-52, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22083642

RESUMO

Paradoxical sleep (PS) is characterized by EEG activation with a disappearance of muscle tone and the occurrence of rapid eye movements (REM) in contrast to slow-wave sleep (SWS, also known as non-REM sleep) identified by the presence of delta waves. Soon after the discovery of PS, it was demonstrated that the structures necessary and sufficient for its genesis are restricted to the brainstem. We review here recent results indicating that brainstem glutamatergic and GABAergic, rather than cholinergic and monoaminergic, neurons play a key role in the genesis of PS. We hypothesize that the entrance to PS from SWS is due to the activation of PS-on glutamatergic neurons localized in the pontine sublaterodorsal tegmental nucleus. The activation of these neurons would be due to a permanent glutamatergic input arising from the lateral and ventrolateral periaqueductal gray (vlPAG) and the removal at the onset of PS of a GABAergic inhibition present during W and SWS. Such inhibition would be coming from PS-off GABAergic neurons localized in the vlPAG and the adjacent deep mesencephalic reticular nucleus. The cessation of activity of these PS-off GABAergic neurons at the onset and during PS would be due to direct projections from intermingled GABAergic PS-on neurons. Activation of PS would depend on the reciprocal interactions between the GABAergic PS-on and PS-off neurons, intrinsic cellular and molecular events, and integration of multiple physiological parameters.


Assuntos
Tronco Encefálico/fisiologia , Sono REM/fisiologia , Animais , Humanos , Modelos Neurológicos , Rede Nervosa/fisiologia
7.
Sleep Med Rev ; 65: 101683, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36096986

RESUMO

Narcolepsy type 1 (NT1) is a rare neurological sleep disorder triggered by postnatal loss of the orexin/hypocretin neuropeptides. Overweight/obesity and precocious puberty are highly prevalent comorbidities of NT1, with a close temporal correlation with disease onset, suggesting a common origin. However, the underlying mechanisms remain unknown and merit further investigation. The main question we address in this review is whether the occurrence of precocious puberty in NT1 is due to the lack of orexin/hypocretin or rather to a wider hypothalamic dysfunction in the context of neuroinflammation, which is likely to accompany the disease given its autoimmune origins. Our analysis suggests that the suspected generalized neuroinflammation of the hypothalamus in NT1 would tend to delay puberty rather than hastening it. In contrast, that the brutal loss of orexin/hypocretin would favor an early reactivation of gonadotropin-releasing hormone (GnRH) secretion during the prepubertal period in vulnerable children, leading to early puberty onset. Orexin/hypocretin replacement could thus be envisaged as a potential treatment for precocious puberty in NT1. Additionally, we put forward an alternative hypothesis regarding the concomitant occurrence of sleepiness, weight gain and early puberty in NT1.


Assuntos
Narcolepsia , Neuropeptídeos , Puberdade Precoce , Criança , Hormônio Liberador de Gonadotropina , Humanos , Doenças Neuroinflamatórias , Orexinas , Puberdade Precoce/complicações
8.
Sleep ; 45(7)2022 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-35429396

RESUMO

Narcolepsy type 1 (NT1) is a rare neurology disorder caused by the loss of orexin/hypocretin neurons. NT1 is characterized by excessive daytime sleepiness, sleep and wake fragmentation, and cataplexy. These symptoms have been equally described in both women and men, although influences of gender and hormonal cycles have been poorly studied. Unfortunately, most studies with NT1 preclinical mouse models, use only male mice to limit potential variations due to the hormonal cycle. Therefore, whether gender and/or hormonal cycles impact the expression of narcoleptic symptoms remains to be determined. To address this question, we analyzed vigilance states and cataplexy in 20 female and 17 male adult orexin knock-out narcoleptic mice, with half of the females being recorded over multiple days. Mice had access to chocolate to encourage the occurrence of cataplectic episodes. A vaginal smear was performed daily in female mice to establish the state of the estrous cycle (EC) of the previous recorded night. We found that vigilance states were more fragmented in males than females, and that females had less paradoxical sleep (p = 0.0315) but more cataplexy (p = 0.0375). Interestingly, sleep and wake features were unchanged across the female EC, but the total amount of cataplexy was doubled during estrus compared to other stages of the cycle (p = 0.001), due to a large increase in the number of cataplexy episodes (p = 0.0002). Altogether these data highlight sex differences in the expression of narcolepsy symptoms in orexin knock-out mice. Notably, cataplexy occurrence was greatly influenced by estrous cycle. Whether it is due to hormonal changes would need to be further explored.


Assuntos
Cataplexia , Narcolepsia , Animais , Cataplexia/diagnóstico , Ciclo Estral , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Narcolepsia/diagnóstico , Narcolepsia/genética , Orexinas/genética , Orexinas/metabolismo , Sono/fisiologia
9.
Brain Pathol ; 32(2): e13027, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34672414

RESUMO

An increased number of histaminergic neurons, identified by labeling histidine-decarboxylase (HDC) its synthesis enzyme, was unexpectedly found in patients with narcolepsy type 1 (NT1). In quest for enlightenment, we evaluate whether an increase in HDC cell number and expression level would be detected in mouse models of the disease, in order to provide proof of concepts reveling possible mechanisms of compensation for the loss of orexin neurons, and/or of induced expression as a consequence of local neuroinflammation, a state that likely accompanies NT1. To further explore the compensatory hypothesis, we also study the noradrenergic wake-promoting system. Immunohistochemistry for HDC, orexin, and melanin-concentrating hormone (MCH) was used to count neurons. Quantitative-PCR of HDC, orexin, MCH, and tyrosine-hydroxylase was performed to evaluate levels of mRNA expression in the hypothalamus or the dorsal pons. Both quantifications were achieved in genetic and neuroinflammatory models of narcolepsy with major orexin impairment, namely the orexin-deficient (Orex-KO) and orexin-hemagglutinin (Orex-HA) mice respectively. The number of HDC neurons and mRNA expression level were unchanged in Orex-KO mice compared to controls. Similarly, we found no change in tyrosine-hydroxylase mRNA expression in the dorsal pons between groups. Further, despite the presence of protracted local neuroinflammation as witnessed by the presence of reactive microglia, we found no change in the number of neurons nor the expression of HDC in Orex-HA mice compared to controls. Importantly, no correlation was found in all conditions between HDC and orexin. Our findings indicate that, in mice, the expression of histamine and noradrenalin, two wake-promoting systems, are not modulated by orexin level whether the lack of orexin is constitutive or induced at adult age, showing thus no compensation. They also show no recruitment of histamine by local neuroinflammation. Further studies will be needed to further define the role of histamine in the pathophysiology of NT1.


Assuntos
Histamina , Narcolepsia , Animais , Histamina/metabolismo , Histidina Descarboxilase/genética , Humanos , Camundongos , Oxigenases de Função Mista , Narcolepsia/genética , Narcolepsia/metabolismo , Orexinas/metabolismo , RNA Mensageiro
10.
Neuron ; 107(3): 397-398, 2020 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-32758443

RESUMO

Memory formation is a dynamic process and sleep is part of it. Consolidation of memories relies on finely orchestrated brain activities occurring during the post-learning sleep period. In this issue of Neuron, Kumar and colleagues provide evidence that the activity of adult-born hippocampal neurons during REM sleep is critical for the consolidation of episodic memory.


Assuntos
Consolidação da Memória , Hipocampo , Neurônios , Sono , Sono REM
11.
Sleep ; 43(7)2020 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-31927578

RESUMO

Identifying the precise neuronal networks activated during paradoxical sleep (PS, also called REM sleep) has been a challenge since its discovery. Similarly, our understanding of the homeostatic mechanisms regulating PS, whether through external modulation by circadian and ultradian drives or via intrinsic homeostatic regulation, is still limited, largely due to interfering factors rendering the investigation difficult. Indeed, none of the studies published so far were able to manipulate PS without significantly altering slow-wave sleep and/or stress level, thus introducing a potential bias in the analyses. With the aim of achieving a better understanding of PS homeostasis, we developed a new method based on automated scoring of vigilance states-using electroencephalogram and electromyogram features-and which involves closed-loop PS deprivation through the induction of cage floor movements when PS is detected. Vigilance states were analyzed during 6 and 48 h of PS deprivation as well as their following recovery periods. Using this new automated methodology, we were able to deprive mice of PS with high efficiency and specificity, for short or longer periods of time, observing no sign of stress (as evaluated by plasma corticosterone level and sleep latency) and requiring no human intervention or environmental changes. We show here that PS can be homeostatically modulated and regulated while no significant changes are induced on slow-wave sleep and wakefulness, with a PS rebound duration depending on the amount of prior PS deficit. We also show that PS interval duration is not correlated with prior PS episode duration in the context of recovery from PS deprivation.


Assuntos
Privação do Sono , Sono REM , Animais , Eletroencefalografia , Homeostase , Camundongos , Sono , Vigília
12.
BMC Genomics ; 9: 418, 2008 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-18796152

RESUMO

BACKGROUND: "Open" transcriptome analysis methods allow to study gene expression without a priori knowledge of the transcript sequences. As of now, SAGE (Serial Analysis of Gene Expression), LongSAGE and MPSS (Massively Parallel Signature Sequencing) are the mostly used methods for "open" transcriptome analysis. Both LongSAGE and MPSS rely on the isolation of 21 pb tag sequences from each transcript. In contrast to LongSAGE, the high throughput sequencing method used in MPSS enables the rapid sequencing of very large libraries containing several millions of tags, allowing deep transcriptome analysis. However, a bias in the complexity of the transcriptome representation obtained by MPSS was recently uncovered. RESULTS: In order to make a deep analysis of mouse hypothalamus transcriptome avoiding the limitation introduced by MPSS, we combined LongSAGE with the Solexa sequencing technology and obtained a library of more than 11 millions of tags. We then compared it to a LongSAGE library of mouse hypothalamus sequenced with the Sanger method. CONCLUSION: We found that Solexa sequencing technology combined with LongSAGE is perfectly suited for deep transcriptome analysis. In contrast to MPSS, it gives a complex representation of transcriptome as reliable as a LongSAGE library sequenced by the Sanger method.


Assuntos
Perfilação da Expressão Gênica/métodos , Análise de Sequência de DNA/métodos , Animais , Biblioteca Gênica , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos
13.
Sleep ; 41(6)2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29522212

RESUMO

Narcolepsy type 1 is a disabling disorder with four primary symptoms: excessive-daytime-sleepiness, cataplexy, hypnagogic hallucinations, and sleep paralysis. The later three symptoms together with a short rapid eye movement (REM) sleep latency have suggested impairment in REM sleep homeostatic regulation with an enhanced propensity for (i.e. tendency to enter) REM sleep. To test this hypothesis, we challenged REM sleep homeostatic regulation in a recognized model of narcolepsy, the orexin knock-out (Orex-KO) mice and their wild-type (WT) littermates. We first performed 48 hr of REM sleep deprivation using the classic small-platforms-over-water method. We found that narcoleptic mice are similarly REM sleep deprived to WT mice. Although they had shorter sleep latency, Orex-KO mice recovered similarly to WT during the following 10 hr of recovery. Interestingly, Orex-KO mice also had cataplexy episodes immediately after REM sleep deprivation, anticipating REM sleep rebound, at a time of day when cataplexy does not occur in baseline condition. We then evaluated REM sleep propensity using our new automated method of deprivation that performs a specific and efficient REM sleep deprivation. We showed that REM sleep propensity is similar during light phase in Orex-KO and WT mice. However, during the dark phase, REM sleep propensity was not suppressed in Orex-KO mice when hypocretin/orexin neuropeptides are normally released. Altogether our data suggest that in addition to the well-known wake-promoting role of hypocretin/orexin, these neuropeptides would also suppress REM sleep. Therefore, hypocretin/orexin deficiency would facilitate the occurrence of REM sleep at any time of day in an opportunistic manner as seen in human narcolepsy.


Assuntos
Homeostase/fisiologia , Narcolepsia/sangue , Narcolepsia/fisiopatologia , Orexinas/sangue , Sono REM/fisiologia , Animais , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Narcolepsia/diagnóstico , Neuropeptídeos/deficiência , Orexinas/deficiência , Privação do Sono/diagnóstico , Privação do Sono/fisiopatologia , Paralisia do Sono/sangue , Paralisia do Sono/diagnóstico , Paralisia do Sono/fisiopatologia
14.
Sleep Med ; 49: 53-63, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30078667

RESUMO

The dorsal raphe nucleus (DRN) through its extensive efferent projections has been implicated in a great variety of physiological and behavioral functions including the regulation of the sleep-wake cycle. This nucleus is composed of five sub-regions defined according to the distribution of its serotonergic (5-HT) neurons. In addition to its heterogeneity in neuronal populations, the DRN contains a great diversity of 5-HT neuronal subtypes identified based on their electrophysiological characteristics, morphology and sub-regional distribution. This suggests that the DRN sub-regions may play different functional roles. Recent studies reported long-range inputs specific to the 5-HT neurons of the DRN; but they did not differentiate whether some inputs were specific to a DRN sub-region, or another region. To fulfill this gap, we have previously described the forebrain afferents to the different sub-regions of the DRN using cholera toxin b subunit and Phaseolus vulgaris-leucoagglutinin, as retrograde and anterograde tracers respectively. In the present work, we provide a detailed map of the brainstem projections to these different sub-regions. We show that if some brainstem structures project homogeneously to all sub-regions, most of the brainstem long-range inputs project in a topographically organized manner onto the DRN and, moreover, that a rich interconnected network is present within the DRN.


Assuntos
Tronco Encefálico/fisiologia , Núcleo Dorsal da Rafe/fisiologia , Vias Neurais/fisiologia , Neurotransmissores/fisiologia , Serotonina/metabolismo , Animais , Toxina da Cólera , Núcleo Dorsal da Rafe/anatomia & histologia , Núcleo Dorsal da Rafe/patologia , França , Imuno-Histoquímica/métodos , Masculino , Vias Neurais/anatomia & histologia , Neurônios/fisiologia , Fito-Hemaglutininas , Ratos , Pesquisa , Vigília/fisiologia
15.
J Comp Neurol ; 505(2): 147-57, 2007 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-17853446

RESUMO

Although the main nodes of the neuronal network that regulate paradoxical sleep (PS), also called rapid eye movement sleep, have been identified in rodents, it still needs to be more thoroughly described. We have recently shown that 58% of a hypothalamic neuronal population, the melanin-concentrating hormone (MCH) neurons, are activated after a PS hypersomnia and that MCH, when injected intracerebroventricularly, induces a dose-dependent increase in PS. This suggests that MCH plays a role in PS regulation. Two subpopulations of MCH neurons have been distinguished neurochemically, one that coexpresses cocaine and amphetamine-regulated transcript (CART) and sends ascending projections to the septum and the hippocampus, the other, the non-CART MCH neurons, send descending projections to the lower brainstem and the spinal cord. In order to better characterize the PS-activated MCH neurons it is interesting to determine whether they belong to the first, the second, or both subgroups. We therefore undertook an MCH, CART, and Fos triple immunolabeling study in PS hypersomniac rats. We showed that the MCH neurons activated during PS are part of both subpopulations since we found CART and non-CART MCH-activated neurons. Based on these results and the literature, we propose that MCH could be involved in memory processes and in the inhibition of muscle tone during PS.


Assuntos
Distúrbios do Sono por Sonolência Excessiva/patologia , Distúrbios do Sono por Sonolência Excessiva/fisiopatologia , Regulação da Expressão Gênica/fisiologia , Hormônios Hipotalâmicos/metabolismo , Melaninas/metabolismo , Neurônios/metabolismo , Hormônios Hipofisários/metabolismo , Animais , Contagem de Células/métodos , Distúrbios do Sono por Sonolência Excessiva/etiologia , Análise de Fourier , Hipotálamo/patologia , Masculino , Proteínas do Tecido Nervoso/metabolismo , Proteínas Oncogênicas v-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Privação do Sono/complicações
16.
Sleep Med Rev ; 32: 85-94, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27083772

RESUMO

The role of gamma-amino butyric acid (GABA) in sleep induction and maintenance is well accepted since most insomnia treatments target GABAa receptors. However, the population(s) of GABAergic neurons involved in the beneficial effect of GABA on sleep remains to be identified. This is not an easy task since GABAergic neurons are widely distributed in all brain structures. A recently growing number of populations of GABAergic neurons have been involved in sleep control. We first review here possible candidates for inducing non-rapid eye movement (NREM) sleep including the GABAergic neurons of the ventrolateral preoptic area, the parafacial zone in the brainstem, the nucleus accumbens and the cortex. We also discuss the role of several populations of GABAergic neurons in rapid eye movement (REM) sleep control. Indeed, it is well accepted that muscle atonia occurring during REM sleep is due to a GABA/glycinergic hyperpolarization of motoneurons. Recent evidence strongly suggests that these neurons are located in the ventral medullary reticular formation. It has also recently been shown that neurons containing the neuropeptide melanin concentrating hormone and GABA located in the lateral hypothalamic area control REM sleep expression. Finally, a population of REM-off GABAergic neurons located in the ventrolateral periaqueductal gray has been shown to gate REM sleep by inhibiting glutamatergic neurons located in the sublaterodorsal tegmental nucleus. In summary, recent data clearly indicate that multiple populations of GABAergic neurons located throughout the brain from the cortex to the medulla oblongata control NREM and REM sleep.


Assuntos
Neurônios GABAérgicos/fisiologia , Sono/fisiologia , Ácido gama-Aminobutírico/fisiologia , Animais , Encéfalo/metabolismo , Humanos , Ratos , Receptores de GABA/fisiologia , Distúrbios do Início e da Manutenção do Sono , Sono REM/fisiologia
17.
CNS Drugs ; 31(10): 821-834, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28940143

RESUMO

Narcolepsy type 1 (NT1) is a rare sleep disorder caused by the very specific loss of hypothalamic hypocretin (Hcrt)/orexin neurons. The exact underlying process leading to this destruction is yet unknown, but indirect evidence strongly supports an autoimmune origin. The association with immune-related genetic factors, in particular the strongest association ever reported in a disease with an allele of a human leukocyte antigen (HLA) gene, and with environmental factors (i.e., the H1N1 influenza infection and vaccination during the pandemic in 2009) are in favor of such a hypothesis. The loss of Hcrt neurons is irreversible, and NT1 is currently an incurable and disabling condition. Patients are managed with symptomatic medication, targeting the main symptoms (excessive daytime sleepiness, cataplexy, disturbed nocturnal sleep), and they require a lifelong treatment. Improved diagnostic tools, together with an increased understanding of the pathogenesis of NT1, may lead to new therapeutic and even preventive interventions. One future treatment could include Hcrt replacement, but this neuropeptide does not cross the blood-brain barrier. However, Hcrt receptor agonists may be promising candidates to treat NT1. Another option is immune-based therapies, administered at disease onset, with already some initiatives to slow down or stop the dysimmune process. Whether immune-based therapy could be beneficial in NT1 remains, however, to be proven.


Assuntos
Doenças Autoimunes/complicações , Doenças Autoimunes/tratamento farmacológico , Narcolepsia/complicações , Narcolepsia/tratamento farmacológico , Animais , Doenças Autoimunes/genética , Causalidade , Modelos Animais de Doenças , Exposição Ambiental/efeitos adversos , Antígenos HLA-A/genética , Humanos , Narcolepsia/etiologia , Narcolepsia/genética
18.
J Physiol Paris ; 100(5-6): 271-83, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17689057

RESUMO

In the middle of the last century, Michel Jouvet discovered paradoxical sleep (PS), a sleep phase paradoxically characterized by cortical activation and rapid eye movements and a muscle atonia. Soon after, he showed that it was still present in "pontine cats" in which all structures rostral to the brainstem have been removed. Later on, it was demonstrated that the pontine peri-locus coeruleus alpha (peri-LCalpha in cats, corresponding to the sublaterodorsal nucleus, SLD, in rats) is responsible for PS onset. It was then proposed that the onset and maintenance of PS is due to a reciprocal inhibitory interaction between neurons presumably cholinergic specifically active during PS localized in this region and monoaminergic neurons. In the last decade, we have tested this hypothesis with our model of head-restrained rats and functional neuroanatomical studies. Our results confirmed that the SLD in rats contains the neurons responsible for the onset and maintenance of PS. They further indicate that (1) these neurons are non-cholinergic possibly glutamatergic neurons, (2) they directly project to the glycinergic premotoneurons localized in the medullary ventral gigantocellular reticular nucleus (GiV), (3) the main neurotransmitter responsible for their inhibition during waking (W) and slow wave sleep (SWS) is GABA rather than monoamines, (4) they are constantly and tonically excited by glutamate and (5) the GABAergic neurons responsible for their tonic inhibition during W and SWS are localized in the deep mesencephalic reticular nucleus (DPMe). We also showed that the tonic inhibition of locus coeruleus (LC) noradrenergic and dorsal raphe (DRN) serotonergic neurons during sleep is due to a tonic GABAergic inhibition by neurons localized in the dorsal paragigantocellular reticular nucleus (DPGi) and the ventrolateral periaqueductal gray (vlPAG). We propose that these GABAergic neurons also inhibit the GABAergic neurons of the DPMe at the onset and during PS and are therefore responsible for the onset and maintenance of PS.


Assuntos
Acetilcolina/metabolismo , Aminas/metabolismo , Ácido Glutâmico/metabolismo , Modelos Biológicos , Sono REM/fisiologia , Ácido gama-Aminobutírico/metabolismo , Animais , Tronco Encefálico/citologia , Tronco Encefálico/fisiologia , Humanos , Redes Neurais de Computação , Vias Neurais/fisiologia , Neurônios/fisiologia
19.
SICOT J ; 2: 33, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27716461

RESUMO

INTRODUCTION: The shoulder arthroplasty brings satisfaction to patients in terms of quality of life and indolence. However whether anatomic implant or reverse, it does not escape from the loosening of the glenoid component. Moreover, optimal implantation is required to ensure the functional outcome without shortening of the arm. The purpose of this study is obtain CT scan evaluation of the glenoid bone stock in order to optimize glenoid component implantation and obtain a reference to determine optimal humeral component placement in case of humeral proximal fracture. MATERIALS AND METHODS: Between 2010 and 2011 we have analyzed 200 intact shoulder's CT. We measured maximal and minimal width in the transverse plane of the glenoid, the distance from the pectoralis major (PM) tendon to the humeral head, the greater tubercle, change of curvature and the anatomical neck. RESULTS: Mean maximum width was 27.4 ± 3.4 mm and mean minimum width was 15.5 ± 2.8 mm. Distances between upper edge of PM tendon to: humeral head, greater tubercle, change of curvature and anatomical neck were respectively: 67.6 ± 9.98 mm, 57.8 ± 10.3 mm, 28.7 ± 9 mm, and 34.2 ± 9.7 mm. CONCLUSION: Our study has produced an assessment of glenoid bone stock for optimal positioning of the glenoid implant but also to obtain a reference to determine the ideal location of the humeral component in the case of proximal humerus fracture.

20.
Sleep ; 38(10): 1537-46, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26085297

RESUMO

STUDY OBJECTIVES: Obstructive sleep apnea (OSA) induces cognitive impairment that involves intermittent hypoxia (IH). Because OSA is recognized as a low-grade systemic inflammatory disease and only some patients develop cognitive deficits, we investigated whether IH-related brain consequences shared similar pathophysiology and required additional factors such as systemic inflammation to develop. DESIGN: Nine-week-old male C57BL/6J mice were exposed to 1 day, 6 or 24 w of IH (alternating 21-5% FiO2 every 30 sec, 8 h/day) or normoxia. Microglial changes were assessed in the functionally distinct dorsal (dH) and ventral (vH) regions of the hippocampus using Iba1 immunolabeling. Then the study concerned dH, as vH only tended to be lately affected. Seven proinflammatory and anti-inflammatory cytokine messenger RNA (mRNA) were assessed at all time points using semiquantitative real-time reverse transcription polymerase chain reaction (RT-PCR). Similar mRNA analysis was performed after 6 w IH or normoxia associated for the past 3 w with repeated intraperitoneal low-dose lipopolysaccharide or saline. MEASUREMENTS AND RESULTS: Chronic (6, 24 w) but not acute IH induced significant microglial changes in dH only, including increased density and morphological features of microglia priming. In dH, acute but not chronic IH increased IL-1ß and RANTES/CCL5 mRNA, whereas the other cytokines remained unchanged. In contrast, chronic IH plus lipopolysaccharide increased interleukin (IL)-6 and IL10 mRNA whereas lipopolysaccharide alone did not affect these cytokines. CONCLUSION: The obstructive sleep apnea component intermittent hypoxia (IH) causes low-grade neuroinflammation in the dorsal hippocampus of mice, including early but transient cytokine elevations, delayed but long-term microglial changes, and cytokine response alterations to lipopolysaccharide inflammatory challenge. These changes may contribute to IH-induced cognitive impairment and pathological brain aging.


Assuntos
Hipocampo/patologia , Hipóxia/complicações , Hipóxia/metabolismo , Inflamação/complicações , Inflamação/patologia , Apneia Obstrutiva do Sono/complicações , Apneia Obstrutiva do Sono/metabolismo , Animais , Quimiocina CCL5/genética , Doença Crônica , Transtornos Cognitivos/complicações , Transtornos Cognitivos/genética , Transtornos Cognitivos/fisiopatologia , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Hipóxia/genética , Hipóxia/patologia , Inflamação/genética , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Interleucinas/genética , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Microglia/patologia , RNA Mensageiro/análise , RNA Mensageiro/genética , Apneia Obstrutiva do Sono/genética , Apneia Obstrutiva do Sono/patologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA