Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Brain ; 145(6): 2018-2030, 2022 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-35552381

RESUMEN

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.


Asunto(s)
Autoinmunidad , Vacunas contra la Influenza , Narcolepsia , Animales , Autoantígenos , Hemaglutininas , Inflamación/complicaciones , Vacunas contra la Influenza/efectos adversos , Interferón gamma , Ratones , Narcolepsia/inducido químicamente , Neuronas , Orexinas , Linfocitos T/inmunología , Vacunación/efectos adversos
2.
PLoS Biol ; 16(10): e2005982, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30307933

RESUMEN

It is crucial to determine whether rapid eye movement (REM) sleep and slow-wave sleep (SWS) (or non-REM sleep), identified in most mammals and birds, also exist in lizards, as they share a common ancestor with these groups. Recently, a study in the bearded dragon (P. vitticeps) reported states analogous to REM and SWS alternating in a surprisingly regular 80-s period, suggesting a common origin of the two sleep states across amniotes. We first confirmed these results in the bearded dragon with deep brain recordings and electro-oculogram (EOG) recordings. Then, to confirm a common origin and more finely characterize sleep in lizards, we developed a multiparametric approach in the tegu lizard, a species never recorded to date. We recorded EOG, electromyogram (EMG), heart rate, and local field potentials (LFPs) and included data on arousal thresholds, sleep deprivation, and pharmacological treatments with fluoxetine, a serotonin reuptake blocker that suppresses REM sleep in mammals. As in the bearded dragon, we demonstrate the existence of two sleep states in tegu lizards. However, no clear periodicity is apparent. The first sleep state (S1 sleep) showed high-amplitude isolated sharp waves, and the second sleep state (S2 sleep) displayed 15-Hz oscillations, isolated ocular movements, and a decrease in heart rate variability and muscle tone compared to S1. Fluoxetine treatment induced a significant decrease in S2 quantities and in the number of sharp waves in S1. Because S2 sleep is characterized by the presence of ocular movements and is inhibited by a serotonin reuptake inhibitor, as is REM sleep in birds and mammals, it might be analogous to this state. However, S2 displays a type of oscillation never previously reported and does not display a desynchronized electroencephalogram (EEG) as is observed in the bearded dragons, mammals, and birds. This suggests that the phenotype of sleep states and possibly their role can differ even between closely related species. Finally, our results suggest a common origin of two sleep states in amniotes. Yet, they also highlight a diversity of sleep phenotypes across lizards, demonstrating that the evolution of sleep states is more complex than previously thought.


Asunto(s)
Lagartos/fisiología , Sueño REM/fisiología , Sueño/fisiología , Animales , Evolución Biológica , Aves/fisiología , Encéfalo , Electroencefalografía/métodos , Electromiografía/métodos , Movimientos Oculares , Fluoxetina/farmacología , Mamíferos/fisiología , Filogenia , Privación de Sueño/fisiopatología , Sueño de Onda Lenta/fisiología
3.
J Sleep Res ; 29(6): e12976, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-31943457

RESUMEN

The cFos immunostaining allowed the identification of multiple populations of neurons involved in the generation of paradoxical sleep. We adopted the transgenic (targeted recombination in active populations) mouse model, which following injection of tamoxifen, allows expression of Cre-dependent reporter constructs (i.e., mCherry) in neurons expressing cFos during waking or paradoxical sleep hypersomnia following automatic paradoxical sleep deprivation. Three groups of mice were subjected to two periods of waking, one period of waking and one of paradoxical sleep hypersomnia, or two periods of paradoxical sleep hypersomnia. A high percentage of double-labelled neurons was observed in the lateral hypothalamic area and zona incerta of two periods of waking and two periods of paradoxical sleep hypersomnia in mice, but not in those of one period of waking and one of paradoxical sleep hypersomnia in animals. Melanin-concentrating hormone neurons in the lateral hypothalamic area and Lhx6+ cells in the zona incerta constituted 5.7 ± 1.5% and 8.8 ± 2.3% of all mCherry+ cells and 20.6 ± 4.8% and 24.6 ± 5.9% of all cFos+ neurons in two periods of paradoxical sleep hypersomnia in animals. In addition, melanin-concentrating hormone cells as well as Lhx6+ neurons rarely expressed mCherry (or cFos) in the waking condition, in contrast to orexin neurons, which constituted approximately 30% of mCherry+ and cFos+ neurons. Our results validate the TRAP methodology and open the way to use it for identifying the neurons activated during waking and paradoxical sleep hypersomnia. Furthermore, they indicate for the first time that Lhx6+ neurons in the zona incerta, like melanin-concentrating hormone cells in the lateral hypothalamic area, are activated during paradoxical sleep hypersomnia but not during waking. These results indicate that Lhx6+ neurons might play a role in the control of paradoxical sleep, like the melanin-concentrating hormone cells.


Asunto(s)
Trastornos de Somnolencia Excesiva/genética , Proteínas con Homeodominio LIM/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Factores de Transcripción/metabolismo , Animales , Modelos Animales de Enfermedad , Masculino , Ratones , Modelos Genéticos , Privación de Sueño/metabolismo
4.
Proc Natl Acad Sci U S A ; 113(39): 10956-61, 2016 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-27621438

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Citotoxicidad Inmunológica , Narcolepsia/inmunología , Narcolepsia/patología , Neuronas/patología , Orexinas/metabolismo , Animales , Autoanticuerpos/metabolismo , Autoantígenos/metabolismo , Comunicación Celular , Hemaglutininas/metabolismo , Hipotálamo/metabolismo , Inflamación/patología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Neuronas/metabolismo , Fenotipo , Linfocitos T Citotóxicos/metabolismo , Células TH1/metabolismo
5.
J Zoo Wildl Med ; 49(2): 291-296, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29900770

RESUMEN

To define a protocol of anesthesia for long-duration invasive surgery in a lizard, eight young adult Argentine tegus ( Salvator merianae) of mean body weight 3.0 kg (interquartile range [IQR] 3.40-2.65) were anesthetized with a mixture of ketamine (K) and medetomidine (M) at 19°C, injected intramuscularly and equally distributed in the four limbs. As the experimental surgery procedure required a prolonged deep anesthesia with a good myorelaxation (between 16 and 21 hr), reinjections were required and reflexes were checked during surgery. Times for anesthetic induction, anesthetic reinjection, and recovery periods were recorded for five different combinations of ketamine-medetomidine: 1) 66 mg/kg K + 100 µg/kg M; 2) 80 mg/kg K + 100 µg/kg M; 3) 100 mg/kg K + 130 µg/kg M; 4) 125 mg/kg K + 200 µg/kg M; and 5) 150 mg/kg K + 200 µg/kg M. The effect on the recovery speed of the postoperative atipamezole injection was also evaluated. The median induction time was 30 (IQR 35-27.5) min with no statistical difference between all the concentrations tested. The first reinjection of half a dose was administered after a mean of 5 hr (5.64 hr, IQR 5.95-4.84) as were the subsequent reinjections of a quarter dose (3.99 hr, IQR 5.98-3.23). Intramuscular administration of the ketamine-medetomidine combination is a simple, rapid, and efficient anesthesia for long-term surgery (>12 hr). A mix of 100 mg/kg ketamine and 200 µg/kg medetomidine, with reinjections every 4 hr of half a dose of the previous injection can maintain a good quality of anesthesia for at least 16 hr. The injection of atipamezole after the surgery reverses the effects of medetomidine and permits a reduction of the recovery period.


Asunto(s)
Anestesia/veterinaria , Anestésicos Disociativos/farmacología , Hipnóticos y Sedantes/farmacología , Ketamina/farmacología , Lagartos/fisiología , Medetomidina/farmacología , Anestésicos Disociativos/administración & dosificación , Animales , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Femenino , Hipnóticos y Sedantes/administración & dosificación , Inyecciones Intramusculares/veterinaria , Ketamina/administración & dosificación , Masculino , Medetomidina/administración & dosificación
6.
Cereb Cortex ; 26(4): 1488-1500, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25585510

RESUMEN

Prolonged rapid-eye-movement (REM) sleep deprivation has long been used to study the role of REM sleep in learning and memory processes. However, this method potentially induces stress and fatigue that may directly affect cognitive functions. Here, by using a short-term and nonstressful REM sleep deprivation (RSD) method we assessed in rats the bidirectional influence of reduced and increased REM sleep amount on hippocampal-dependent emotional memory and plasticity. Our results indicate that 4 h RSD impaired consolidation of contextual fear conditioning (CFC) and induction of long-term potentiation (LTP), while decreasing density of Egr1/Zif268-expressing neurons in the CA1 region of the dorsal hippocampus. LTP and Egr1 expression were not affected in ventral CA1. Conversely, an increase in REM sleep restores and further facilitates CFC consolidation and LTP induction, and also increases Egr1 expression in dorsal CA1. Moreover, CFC consolidation, Egr1 neuron density, and LTP amplitude in dorsal CA1 show a positive correlation with REM sleep amount. Altogether, these results indicate that mild changes in REM sleep amount bidirectionally affect memory and synaptic plasticity mechanisms occurring in the CA1 area of the dorsal hippocampus.


Asunto(s)
Emociones/fisiología , Hipocampo/fisiopatología , Potenciación a Largo Plazo , Consolidación de la Memoria/fisiología , Privación de Sueño/fisiopatología , Sueño REM , Animales , Condicionamiento Clásico/fisiología , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Miedo/fisiología , Hipocampo/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley
7.
J Neurosci ; 35(27): 9900-11, 2015 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-26156991

RESUMEN

Sleep-active neurons located in the ventrolateral preoptic nucleus (VLPO) play a crucial role in the induction and maintenance of slow-wave sleep (SWS). However, the cellular and molecular mechanisms responsible for their activation at sleep onset remain poorly understood. Here, we test the hypothesis that a rise in extracellular glucose concentration in the VLPO can promote sleep by increasing the activity of sleep-promoting VLPO neurons. We find that infusion of a glucose concentration into the VLPO of mice promotes SWS and increases the density of c-Fos-labeled neurons selectively in the VLPO. Moreover, we show in patch-clamp recordings from brain slices that VLPO neurons exhibiting properties of sleep-promoting neurons are selectively excited by glucose within physiological range. This glucose-induced excitation implies the catabolism of glucose, leading to a closure of ATP-sensitive potassium (KATP) channels. The extracellular glucose concentration monitors the gating of KATP channels of sleep-promoting neurons, highlighting that these neurons can adapt their excitability according to the extracellular energy status. Together, these results provide evidence that glucose may participate in the mechanisms of SWS promotion and/or consolidation. SIGNIFICANCE STATEMENT: Although the brain circuitry underlying vigilance states is well described, the molecular mechanisms responsible for sleep onset remain largely unknown. Combining in vitro and in vivo experiments, we demonstrate that glucose likely contributes to sleep onset facilitation by increasing the excitability of sleep-promoting neurons in the ventrolateral preoptic nucleus (VLPO). We find here that these neurons integrate energetic signals such as ambient glucose directly to regulate vigilance states accordingly. Glucose-induced excitation of sleep-promoting VLPO neurons should therefore be involved in the drowsiness that one feels after a high-sugar meal. This novel mechanism regulating the activity of VLPO neurons reinforces the fundamental and intimate link between sleep and metabolism.


Asunto(s)
Glucosa/farmacología , Neuronas/efectos de los fármacos , Área Preóptica/citología , Área Preóptica/metabolismo , Sueño/efectos de los fármacos , Edulcorantes/farmacología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Agonistas alfa-Adrenérgicos/farmacología , Animales , Ondas Encefálicas/efectos de los fármacos , Ácidos Cumáricos/farmacología , Desoxiglucosa/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Transportador de Glucosa de Tipo 3/genética , Transportador de Glucosa de Tipo 3/metabolismo , Técnicas In Vitro , Masculino , Moduladores del Transporte de Membrana/farmacología , Ratones , Ratones Endogámicos C57BL , Norepinefrina/farmacología , Proteínas Proto-Oncogénicas c-fos/metabolismo
8.
J Sleep Res ; 24(3): 309-19, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25524602

RESUMEN

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.


Asunto(s)
Hormonas Hipotalámicas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Melaninas/metabolismo , Neuronas/fisiología , Neuropéptidos/metabolismo , Hormonas Hipofisarias/metabolismo , Privación de Sueño/fisiopatología , Sueño REM/fisiología , Agua , Animales , Atención/fisiología , Corticosterona/metabolismo , Homeostasis , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Orexinas , Polisomnografía , Ratas , Sueño/fisiología , Factores de Tiempo , Vigilia/fisiología
9.
Neuropsychopharmacology ; 49(7): 1129-1139, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38326457

RESUMEN

Treatments are only partially effective in major depressive disorders (MDD) but no biomarker exists to predict symptom improvement in patients. Animal models are essential tools in the development of antidepressant medications, but while recent genetic studies have demonstrated the polygenic contribution to MDD, current models are limited to either mimic the effect of a single gene or environmental factor. We developed in the past a model of depressive-like behaviors in mice (H/Rouen), using selective breeding based on behavioral reaction after an acute mild stress in the tail suspension test. Here, we propose a new mouse model of depression (H-TST) generated from a more complex genetic background and based on the same selection process. We first demonstrated that H/Rouen and H-TST mice had similar phenotypes and were more sensitive to glutamate-related antidepressant medications than selective serotonin reuptake inhibitors. We then conducted an exome sequencing on the two mouse models and showed that they had damaging variants in 174 identical genes, which have also been associated with MDD in humans. Among these genes, we showed a higher expression level of Tmem161b in brain and blood of our two mouse models. Changes in TMEM161B expression level was also observed in blood of MDD patients when compared with controls, and after 8-week treatment with duloxetine, mainly in good responders to treatment. Altogether, our results introduce H/Rouen and H-TST as the two first polygenic animal models of MDD and demonstrate their ability to identify biomarkers of the disease and to develop rapid and effective antidepressant medications.


Asunto(s)
Antidepresivos , Biomarcadores , Trastorno Depresivo Mayor , Modelos Animales de Enfermedad , Herencia Multifactorial , Trastorno Depresivo Mayor/genética , Trastorno Depresivo Mayor/tratamiento farmacológico , Animales , Humanos , Ratones , Masculino , Antidepresivos/farmacología , Antidepresivos/uso terapéutico , Biomarcadores/sangre , Femenino , Adulto , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Persona de Mediana Edad , Encéfalo/metabolismo
10.
J Neurosci ; 32(47): 16763-74, 2012 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-23175830

RESUMEN

It has recently been shown that the ventrolateral part of the periaqueductal gray (VLPAG) and the adjacent dorsal deep mesencephalic nucleus (dDpMe) contain GABAergic neurons gating paradoxical sleep (PS) onset by means of their projection to the glutamatergic PS-on neurons of the sublaterodorsal tegmental nucleus (SLD). To determine the mechanisms responsible for the cessation of activity of these GABAergic PS-off neurons at the onset and during PS, we combined the immunostaining of c-FOS, a marker of neuronal activation, with cholera toxin b subunit (CTb) retrograde tracing from the VLPAG/dDpMe in three groups of rats (control, PS deprived, and PS hypersomniac). We found that the lateral hypothalamic area (LH) is the only brain structure containing a very large number of neurons activated during PS hypersomnia and projecting to the VLPAG/dDpMe. We further demonstrated that 44% of these neurons express the neuropeptide melanin concentrating hormone (MCH). We then showed that bilateral injections in the LH of two inhibitory compounds, clonidine (an α-2 adrenergic agonist) and muscimol (a GABAa agonist) induce an inhibition of PS. Furthermore, after muscimol injections in the LH, the VLPAG/dDpMe contained a large number of activated neurons, mostly GABAergic, and projecting to the SLD. Altogether, our results indicate for the first time that the activation of a population of LH neurons, in part MCH containing, is necessary for PS to occur. Furthermore, our results strongly suggest that these neurons trigger PS by means of their inhibitory projection to the PS-off GABAergic neurons located in the VLPAG/dDpMe.


Asunto(s)
Tronco Encefálico/fisiología , Neuronas GABAérgicas/fisiología , Área Hipotalámica Lateral/fisiología , Vías Nerviosas/fisiología , Sueño REM/fisiología , Animales , Tronco Encefálico/citología , Tronco Encefálico/efectos de los fármacos , Electroencefalografía/efectos de los fármacos , Electromiografía/efectos de los fármacos , Neuronas GABAérgicas/efectos de los fármacos , Glutamato Descarboxilasa/metabolismo , Área Hipotalámica Lateral/efectos de los fármacos , Inmunohistoquímica , Hibridación in Situ , Masculino , Vías Nerviosas/efectos de los fármacos , Neurotensina/metabolismo , Polisomnografía , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley , Sueño REM/efectos de los fármacos , Fijación del Tejido
11.
Sleep ; 45(7)2022 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-35429396

RESUMEN

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.


Asunto(s)
Cataplejía , Narcolepsia , Animales , Cataplejía/diagnóstico , Ciclo Estral , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Narcolepsia/diagnóstico , Narcolepsia/genética , Orexinas/genética , Orexinas/metabolismo , Sueño/fisiología
12.
Sleep ; 44(12)2021 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-34245290

RESUMEN

STUDY OBJECTIVES: Determine whether in the hippocampus and the supramammillary nucleus (SuM) the same neurons are reactivated when mice are exposed 1 week apart to two periods of wakefulness (W-W), paradoxical sleep rebound (PSR-PSR) or a period of W followed by a period of PSR (W-PSR). METHODS: We combined the innovative TRAP2 mice method in which neurons expressing cFos permanently express tdTomato after tamoxifen injection with cFos immunohistochemistry. RESULTS: We found out that a large number of tdTomato+ and cFos+ cells are localized in the dentate gyrus (DG) after PSR and W while CA1 and CA3 contained both types of neurons only after W. The number of cFos+ cells in the infrapyramidal but not the suprapyramidal blade of the DG was positively correlated with the amount of PS. In addition, we did not find double-labeled cells in the DG whatever the group of mice. In contrast, a high percentage of CA1 neurons were double-labeled in W-W mice. Finally, in the supramammillary nucleus, a large number of cells were double-labeled in W-W, PSR-PSR but not in W-PSR mice. CONCLUSIONS: Altogether, our results are the first to show that different neurons are activated during W and PS in the supramammillary nucleus and the hippocampus. Further, we showed for the first time that granule cells of the infrapyramidal blade of the DG are activated during PS but not during W. Further experiments are now needed to determine whether these granule cells belong to memory engrams inducing memory reactivation during PS.


Asunto(s)
Trastornos de Somnolencia Excesiva , Sueño REM , Animales , Giro Dentado/fisiología , Ratones , Neuronas/fisiología , Sueño REM/fisiología , Vigilia
13.
Biochem Pharmacol ; 191: 114514, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33713640

RESUMEN

Michel Jouvet proposed in 1959 that REM sleep is a paradoxical state since it was characterized by the association of a cortical activation similar to wakefulness (W) with muscle atonia. Recently, we showed using cFos as a marker of activity that cortical activation during paradoxical sleep (PS) was limited to a few limbic cortical structures in contrast to W during which all cortices were strongly activated. However, we were not able to demonstrate whether the same neurons are activated during PS and W and to rule out that the activation observed was not linked with stress induced by the flowerpot method of PS deprivation. In the present study, we answered to these two questions by combining tdTomato and cFos immunostaining in the innovative TRAP2 transgenic mice exposed one week apart to two periods of W (W-W mice), PS rebound (PSR-PSR) or a period of W followed by a period of PSR (W-PSR mice). Using such method, we showed that different neurons are activated during W and PSR in the anterior cingulate (ACA) and rostral and caudal retrosplenial (rRSP and cRSP) cortices as well as the claustrum (CLA) previously shown to contain a large number of activated neurons after PSR. Further, the distribution of the neurons during PSR in the rRSP and cRSP was limited to the superficial layers while it was widespread across all layers during W. Our results clearly show at the cellular level that PS and W are two completely different states in term of neocortical activation.


Asunto(s)
Claustro/fisiología , Trastornos de Somnolencia Excesiva/fisiopatología , Giro del Cíngulo/fisiología , Neuronas/fisiología , Sueño REM/fisiología , Vigilia/fisiología , Animales , Claustro/citología , Trastornos de Somnolencia Excesiva/genética , Trastornos de Somnolencia Excesiva/patología , Femenino , Giro del Cíngulo/citología , Masculino , Ratones , Ratones Transgénicos , Polisomnografía/métodos
14.
J Neurochem ; 113(2): 329-40, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20050979

RESUMEN

It is now established that the development of the CNS requires equilibrium between cell survival and apoptosis. Pituitary adenylate cyclase-activating polypeptide (PACAP) exerts a powerful protective effect on cerebellar granule cells by inhibiting the caspase 3. In contrast, Fas ligand (FasL) plays an essential role during ontogenesis in eliminating supernumerary neurons by apoptosis. To determine if PACAP and FasL interact during cerebellar development, we characterized the effects of these factors on cerebellar morphogenesis and caspase 3 activity in PACAP+/+ and PACAP-/- mice. First, we demonstrated in vivo that PACAP is able to reverse the diminution of internal granule cell layer thickness induced by FasL in PACAP+/+ and PACAP-/- mice. Second, ex vivo and immunohistochemical studies revealed that interaction between FasL and PACAP occurs through the caspase 3 activity. Third, behavioural study showed a significant difference for the PACAP + FasL group in the righting reflex test at P8 which does not persist at P60. Finally, a time course study revealed that the pro-apoptotic effect of FasL characterized at P8 was followed by a progressive compensatory mechanism in caspase 3 activity and bromodeoxyuridine incorporation. These data suggest that PACAP and FasL interact during cerebellar development to control apoptosis of granule cells and may affect some motor cerebellar functions.


Asunto(s)
Conducta Animal/fisiología , Cerebelo/citología , Cerebelo/crecimiento & desarrollo , Proteína Ligando Fas/metabolismo , Neuronas/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Animales , Animales Recién Nacidos , Conducta Animal/efectos de los fármacos , Bromodesoxiuridina/metabolismo , Caspasa 3/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Proliferación Celular/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Cerebelo/efectos de los fármacos , Proteína Ligando Fas/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiología , Neuronas/efectos de los fármacos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/deficiencia , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Desempeño Psicomotor/efectos de los fármacos , ARN Mensajero/metabolismo , Reflejo/efectos de los fármacos , Estadísticas no Paramétricas , Factores de Tiempo
15.
Sleep ; 43(7)2020 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-31927578

RESUMEN

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.


Asunto(s)
Privación de Sueño , Sueño REM , Animales , Electroencefalografía , Homeostasis , Ratones , Sueño , Vigilia
16.
J Neurosci ; 28(26): 6616-26, 2008 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-18579734

RESUMEN

An increase in circulating catecholamine levels represents one of the mechanisms whereby organisms cope with stress. In the periphery, catecholamines mainly originate from the sympathoadrenal system. As we reported, in addition to the central control through cholinergic innervation, a local gap junction-delineated route between adrenal chromaffin cells contributes to catecholamine exocytosis. Here, we investigated whether this intercellular communication is modified when the hormonal demand is increased as observed during cold stress. Our results show that in cold exposed rats, gap-junctional communication undergoes a functional plasticity, as evidenced by an increased number of dye-coupled cells. Of a physiological interest is that this upregulation of gap-junctional coupling results in the appearance of a robust electrical coupling between chromaffin cells that allows the transmission of action potentials between coupled cells. This enhancement of gap-junctional communication parallels an increase in expression levels of connexin36 (Cx36) and connexin43 (Cx43) proteins. Both transcriptional and posttranslational mechanisms are involved because Cx36 transcripts are increased in stressed rats and the expression of the scaffolding protein zonula occludens-1, known to interact with both Cx36 and Cx43, is also upregulated. Consistent with an upregulated coupling extent in stressed rats, the cytosolic Ca(2+) concentration rises triggered in a single cell by an iontophoretic application of nicotine occur simultaneously in several neighboring cells. These results describe for the first time a functional plasticity of junctional coupling between adult chromaffin cells that should be crucial for adaptation to stress or sensitization to subsequent stressors.


Asunto(s)
Médula Suprarrenal/metabolismo , Catecolaminas/metabolismo , Comunicación Celular/fisiología , Células Cromafines/metabolismo , Uniones Comunicantes/metabolismo , Estrés Psicológico/metabolismo , Potenciales de Acción/fisiología , Médula Suprarrenal/ultraestructura , Animales , Calcio/metabolismo , Señalización del Calcio/fisiología , Células Cromafines/ultraestructura , Frío/efectos adversos , Conexina 43/genética , Conexina 43/metabolismo , Conexinas/genética , Conexinas/metabolismo , Uniones Comunicantes/ultraestructura , Masculino , Potenciales de la Membrana/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Plasticidad Neuronal/fisiología , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Ratas , Ratas Wistar , Estrés Psicológico/fisiopatología , Regulación hacia Arriba/fisiología , Proteína de la Zonula Occludens-1 , Proteína delta-6 de Union Comunicante
17.
J Neurosci Methods ; 316: 103-116, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30189286

RESUMEN

BACKGROUND: Sleep is an inactive state of reduced environmental awareness shared by all animals. When compared to wakefulness, sleep behavior is associated with changes in physiology and brain activity. The nature of these changes varies considerably across species, and therefore is a rich resource for gaining insight into the evolution and functions of sleep. A major obstacle to capitalizing on this resource is the lack of a small device capable of recording multiple biological parameters for extended periods of time both in the laboratory and the field. NEW METHOD: ONEIROS is a new tool designed for conducting sleep research on small, freely moving animals. The miniature, standalone system is capable of recording up to 26 electrophysiological signals (electroencephalogram, electromyogram, electrooculogram, electrocardiogram), metabolic (3 temperature channels) and behavior via an accelerometer for several days. In addition, the device is equipped with a vibrating motor which can be used to assess arousal thresholds and to disrupt sleep. The system is available in telemetric or data-logger configuration useable in the field. RESULTS: To demonstrate the efficacy of this tool, we simultaneously recorded for the first time, electroencephalogram, hippocampal local field potential, electromyogram, electrooculogram, brain, body and ambient temperature, and 3D accelerometry. We also deprived rats of paradoxical sleep by triggering the vibrating motor after online recognition of the state. Finally, by successfully recording a pigeon in an 8 m3 aviary in a social context with the device in the logger configuration, we demonstrate the feasibility of using the device in the field.


Asunto(s)
Acelerometría/instrumentación , Electrocardiografía/instrumentación , Electromiografía/instrumentación , Electrooculografía/instrumentación , Monitoreo Fisiológico/instrumentación , Privación de Sueño/fisiopatología , Sueño/fisiología , Telemetría/instrumentación , Acelerometría/métodos , Animales , Electrocardiografía/métodos , Electromiografía/métodos , Electrooculografía/métodos , Masculino , Monitoreo Fisiológico/métodos , Monitorización Neurofisiológica/instrumentación , Monitorización Neurofisiológica/métodos , Ratas , Ratas Sprague-Dawley , Sueño REM/fisiología , Telemetría/métodos
18.
Sleep ; 41(6)2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29522212

RESUMEN

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.


Asunto(s)
Homeostasis/fisiología , Narcolepsia/sangre , Narcolepsia/fisiopatología , Orexinas/sangre , Sueño REM/fisiología , Animales , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Narcolepsia/diagnóstico , Neuropéptidos/deficiencia , Orexinas/deficiencia , Privación de Sueño/diagnóstico , Privación de Sueño/fisiopatología , Parálisis del Sueño/sangre , Parálisis del Sueño/diagnóstico , Parálisis del Sueño/fisiopatología
19.
Nat Commun ; 9(1): 504, 2018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29402935

RESUMEN

Despite decades of research, there is a persistent debate regarding the localization of GABA/glycine neurons responsible for hyperpolarizing somatic motoneurons during paradoxical (or REM) sleep (PS), resulting in the loss of muscle tone during this sleep state. Combining complementary neuroanatomical approaches in rats, we first show that these inhibitory neurons are localized within the ventromedial medulla (vmM) rather than within the spinal cord. We then demonstrate their functional role in PS expression through local injections of adeno-associated virus carrying specific short-hairpin RNA in order to chronically impair inhibitory neurotransmission from vmM. After such selective genetic inactivation, rats display PS without atonia associated with abnormal and violent motor activity, concomitant with a small reduction of daily PS quantity. These symptoms closely mimic human REM sleep behavior disorder (RBD), a prodromal parasomnia of synucleinopathies. Our findings demonstrate the crucial role of GABA/glycine inhibitory vmM neurons in muscle atonia during PS and highlight a candidate brain region that can be susceptible to α-synuclein-dependent degeneration in RBD patients.


Asunto(s)
Bulbo Raquídeo/fisiología , Neuronas/fisiología , Sueño REM/fisiología , Animales , Técnicas de Silenciamiento del Gen , Glicina/metabolismo , Masculino , Bulbo Raquídeo/citología , Hipotonía Muscular/fisiopatología , Polisomnografía , Proteínas Proto-Oncogénicas c-fos/metabolismo , Trastorno de la Conducta del Sueño REM/fisiopatología , Ratas Sprague-Dawley , Transmisión Sináptica/genética , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/genética , Ácido gamma-Aminobutírico/metabolismo
20.
Behav Brain Res ; 298(Pt B): 100-10, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26529469

RESUMEN

Recent reports support a key role of tuberal hypothalamic neurons secreting melanin concentrating-hormone (MCH) in the promotion of Paradoxical Sleep (PS). Controversies remain concerning their concomitant involvement in Slow-Wave Sleep (SWS). We studied the effects of their selective loss achieved by an Ataxin 3-mediated ablation strategy to decipher the contribution of MCH neurons to SWS and/or PS. Polysomnographic recordings were performed on male adult transgenic mice expressing Ataxin-3 transgene within MCH neurons (MCH(Atax)) and their wild-type littermates (MCH(WT)) bred on two genetic backgrounds (FVB/N and C57BL/6). Compared to MCH(WT) mice, MCH(Atax) mice were characterized by a significant drop in MCH mRNAs (-70%), a partial loss of MCH-immunoreactive neurons (-30%) and a marked reduction in brain density of MCH-immunoreactive fibers. Under basal condition, such MCH(Atax) mice exhibited higher PS amounts during the light period and a pronounced SWS fragmentation without any modification of SWS quantities. Moreover, SWS and PS rebounds following 4-h total sleep deprivation were quantitatively similar in MCH(Atax)vs. MCH(WT) mice. Additionally, MCH(Atax) mice were unable to consolidate SWS and increase slow-wave activity (SWA) in response to this homeostatic challenge as observed in MCH(WT) littermates. Here, we show that the partial loss of MCH neurons is sufficient to disturb the fine-tuning of sleep. Our data provided new insights into their contribution to subtle process managing SWS quality and its efficiency rather than SWS quantities, as evidenced by the deleterious impact on two powerful markers of sleep depth, i.e., SWS consolidation/fragmentation and SWA intensity under basal condition and under high sleep pressure.


Asunto(s)
Encéfalo/fisiopatología , Homeostasis/fisiología , Hormonas Hipotalámicas/metabolismo , Melaninas/metabolismo , Neuronas/fisiología , Hormonas Hipofisarias/metabolismo , Sueño/fisiología , Animales , Encéfalo/patología , Recuento de Células , Inmunohistoquímica , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/patología , Polisomnografía , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA