Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Ann Neurol ; 88(3): 626-630, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32533727

RESUMO

In March 2020, we treated a cohort of 26 critically ill hospitalized SARS-CoV-2-infected patients who underwent electroencephalography to assess unexplained altered mental status, loss of consciousness, or poor arousal and responsiveness. Of the 26 patients studied, 5 patients had electroencephalograms that showed periodic discharges consisting of high-amplitude frontal monomorphic delta waves with absence of epileptic activity. These findings may suggest central nervous system injury potentially related to COVID-19 in these patients. ANN NEUROL 2020;88:626-630.


Assuntos
Encefalopatias/fisiopatologia , Encefalopatias/virologia , COVID-19/complicações , COVID-19/fisiopatologia , Idoso , Encéfalo/fisiopatologia , Estado Terminal , Eletroencefalografia , Humanos , Masculino , Pessoa de Meia-Idade
2.
Genes Dev ; 26(24): 2780-801, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23222102

RESUMO

The FoxO family of transcription factors is known to slow aging downstream from the insulin/IGF (insulin-like growth factor) signaling pathway. The most recently discovered FoxO isoform in mammals, FoxO6, is highly enriched in the adult hippocampus. However, the importance of FoxO factors in cognition is largely unknown. Here we generated mice lacking FoxO6 and found that these mice display normal learning but impaired memory consolidation in contextual fear conditioning and novel object recognition. Using stereotactic injection of viruses into the hippocampus of adult wild-type mice, we found that FoxO6 activity in the adult hippocampus is required for memory consolidation. Genome-wide approaches revealed that FoxO6 regulates a program of genes involved in synaptic function upon learning in the hippocampus. Consistently, FoxO6 deficiency results in decreased dendritic spine density in hippocampal neurons in vitro and in vivo. Thus, FoxO6 may promote memory consolidation by regulating a program coordinating neuronal connectivity in the hippocampus, which could have important implications for physiological and pathological age-dependent decline in memory.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Memória/fisiologia , Animais , Contagem de Células , Células Cultivadas , Espinhas Dendríticas/genética , Espinhas Dendríticas/metabolismo , Fatores de Transcrição Forkhead/genética , Deleção de Genes , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Hipocampo/citologia , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Regulação Miogênica/metabolismo , Sinapses/genética , Sinapses/metabolismo
3.
Neurobiol Learn Mem ; 140: 11-16, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28215510

RESUMO

Down syndrome (DS) is a common genetic cause of intellectual disability yet no pro-cognitive drug therapies are approved for human use. Mechanistic studies in a mouse model of DS (Ts65Dn mice) demonstrate that impaired cognitive function is due to excessive neuronal inhibitory tone. These deficits are normalized by chronic, short-term low doses of GABAA receptor (GABAAR) antagonists in adult animals, but none of the compounds investigated are approved for human use. We explored the therapeutic potential of flumazenil (FLUM), a GABAAR antagonist working at the benzodiazepine binding site that has FDA approval. Long-term memory was assessed by the Novel Object Recognition (NOR) testing in Ts65Dn mice after acute or short-term chronic treatment with FLUM. Short-term, low, chronic dose regimens of FLUM elicit long-lasting (>1week) normalization of cognitive function in both young and aged mice. FLUM at low dosages produces long lasting cognitive improvements and has the potential of fulfilling an unmet therapeutic need in DS.


Assuntos
Síndrome de Down/tratamento farmacológico , Flumazenil/uso terapêutico , Moduladores GABAérgicos/uso terapêutico , Transtornos da Memória/tratamento farmacológico , Memória de Longo Prazo/efeitos dos fármacos , Animais , Cognição/efeitos dos fármacos , Modelos Animais de Doenças , Síndrome de Down/genética , Flumazenil/farmacologia , Moduladores GABAérgicos/farmacologia , Masculino , Camundongos
4.
Brain ; 139(Pt 7): 2063-81, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27190010

RESUMO

Identifying preventive targets for Alzheimer's disease is a central challenge of modern medicine. Non-steroidal anti-inflammatory drugs, which inhibit the cyclooxygenase enzymes COX-1 and COX-2, reduce the risk of developing Alzheimer's disease in normal ageing populations. This preventive effect coincides with an extended preclinical phase that spans years to decades before onset of cognitive decline. In the brain, COX-2 is induced in neurons in response to excitatory synaptic activity and in glial cells in response to inflammation. To identify mechanisms underlying prevention of cognitive decline by anti-inflammatory drugs, we first identified an early object memory deficit in APPSwe-PS1ΔE9 mice that preceded previously identified spatial memory deficits in this model. We modelled prevention of this memory deficit with ibuprofen, and found that ibuprofen prevented memory impairment without producing any measurable changes in amyloid-ß accumulation or glial inflammation. Instead, ibuprofen modulated hippocampal gene expression in pathways involved in neuronal plasticity and increased levels of norepinephrine and dopamine. The gene most highly downregulated by ibuprofen was neuronal tryptophan 2,3-dioxygenase (Tdo2), which encodes an enzyme that metabolizes tryptophan to kynurenine. TDO2 expression was increased by neuronal COX-2 activity, and overexpression of hippocampal TDO2 produced behavioural deficits. Moreover, pharmacological TDO2 inhibition prevented behavioural deficits in APPSwe-PS1ΔE9 mice. Taken together, these data demonstrate broad effects of cyclooxygenase inhibition on multiple neuronal pathways that counteract the neurotoxic effects of early accumulating amyloid-ß oligomers.


Assuntos
Doença de Alzheimer/prevenção & controle , Expressão Gênica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Transtornos da Memória/prevenção & controle , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase , Modelos Animais de Doenças , Regulação para Baixo , Eletroencefalografia , Ibuprofeno , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Reconhecimento Psicológico/efeitos dos fármacos , Triptofano Oxigenase/efeitos dos fármacos
5.
Neurobiol Learn Mem ; 116: 162-71, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25463650

RESUMO

Down syndrome (DS) has an incidence of about 1/700 births, and is therefore the most common cause of cognitive and behavioral impairments in children. Recent studies on mouse models of DS indicate that a number of pharmacotherapies could be beneficial for restoring cognitive abilities in individuals with DS. Attention deficits that are present in DS account in part for learning and memory deficiencies yet have been scarcely studied in corresponding models. Investigations of this relevant group of behaviors is more difficult in mouse models because of the difficulty in homologizing mouse and human behaviors and because standard laboratory environments do not always elicit behaviors of interest. Here we characterize nest building as a goal-directed behavior that is seriously impaired in young Ts65Dn mice, a genetic model of DS. We believe this impairment may reflect in part attention deficits, and we investigate the physiological, genetic, and pharmacological factors influencing its expression. Nesting behavior in young Ts65Dn mice was severely impaired when the animals were placed in a novel environment. But this context-dependent impairment was transient and reversible. The genetic determinants of this deficiency are restricted to a ∼100 gene segment on the murine chromosome 16. Nest building behavior is a highly integrated phenotypic trait that relies in part on limbic circuitry and on the frontal cortex in relation to cognitive and attention processes. We show that both serotonin content and 5HT2a receptors are increased in the frontal cortex of Ts65Dn mice and that pharmacological blockage of 5HT2a receptors in Ts65Dn mice rescues their context dependent nest building impairment. We propose that the nest-building trait could represent a marker of attention related deficits in DS models and could be of value in designing pharmacotherapies for this specific aspect of DS. 5HT2a modulation may improve goal-directed behavior in DS.


Assuntos
Transtornos Cognitivos/fisiopatologia , Síndrome de Down/fisiopatologia , Comportamento de Nidação/fisiologia , Receptor 5-HT2A de Serotonina/metabolismo , Antagonistas do Receptor 5-HT2 de Serotonina/farmacologia , Animais , Transtornos Cognitivos/genética , Transtornos Cognitivos/metabolismo , Modelos Animais de Doenças , Síndrome de Down/genética , Síndrome de Down/metabolismo , Expressão Gênica , Camundongos , Comportamento de Nidação/efeitos dos fármacos , Fenótipo , Receptor 5-HT2A de Serotonina/genética , Risperidona/farmacologia
6.
Proc Natl Acad Sci U S A ; 108(32): 13305-10, 2011 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-21788501

RESUMO

Memory consolidation has been proposed as a function of sleep. However, sleep is a complex phenomenon characterized by several features including duration, intensity, and continuity. Sleep continuity is disrupted in different neurological and psychiatric conditions, many of which are accompanied by memory deficits. This finding has raised the question of whether the continuity of sleep is important for memory consolidation. However, current techniques used in sleep research cannot manipulate a single sleep feature while maintaining the others constant. Here, we introduce the use of optogenetics to investigate the role of sleep continuity in memory consolidation. We optogenetically targeted hypocretin/orexin neurons, which play a key role in arousal processes. We used optogenetics to activate these neurons at different intervals in behaving mice and were able to fragment sleep without affecting its overall amount or intensity. Fragmenting sleep after the learning phase of the novel object recognition (NOR) task significantly decreased the performance of mice on the subsequent day, but memory was unaffected if the average duration of sleep episodes was maintained at 62-73% of normal. These findings demonstrate the use of optogenetic activation of arousal-related nuclei as a way to systematically manipulate a specific feature of sleep. We conclude that regardless of the total amount of sleep or sleep intensity, a minimal unit of uninterrupted sleep is crucial for memory consolidation.


Assuntos
Memória/fisiologia , Privação do Sono/fisiopatologia , Sono/genética , Sono/efeitos da radiação , Animais , Eletroencefalografia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Memória/efeitos da radiação , Camundongos , Neurônios/metabolismo , Neurônios/patologia , Neurônios/efeitos da radiação , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Orexinas , Estimulação Física , Privação do Sono/complicações , Privação do Sono/genética , Sono REM/fisiologia , Sono REM/efeitos da radiação , Estresse Psicológico/complicações , Estresse Psicológico/fisiopatologia , Análise e Desempenho de Tarefas , Fatores de Tempo
7.
Eur J Neurosci ; 37(11): 1727-36, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23510299

RESUMO

Light exerts a direct effect on sleep and wakefulness in nocturnal and diurnal animals, with a light pulse during the dark phase suppressing locomotor activity and promoting sleep in the former. In the present study, we investigated this direct effect of light on various sleep parameters by exposing mice to a broad range of illuminances (0.2-200 µW/cm(2) ; equivalent to 1-1000 lux) for 1 h during the dark phase (zeitgeber time 13-14). Fitting the data with a three-parameter log model indicated that ~0.1 µW/cm(2) can generate half the sleep response observed at 200 µW/cm(2). We observed decreases in total sleep time during the 1 h following the end of the light pulse. Light reduced the latency to sleep from ~30 min in darkness (baseline) to ~10 min at the highest intensity, although this effect was invariant across the light intensities used. We then assessed the role of melanopsin during the rapid transition from wakefulness to sleep at the onset of a light pulse and the maintenance of sleep with a 6-h 20 µW/cm(2) light pulse. Even though the melanopsin knockout mice had robust induction of sleep (~35 min) during the first hour of the pulse, it was not maintained. Total sleep decreased by almost 65% by the third hour in comparison with the first hour of the pulse in mice lacking melanopsin, whereas only an 8% decrease was observed in wild-type mice. Collectively, our findings highlight the selective effects of light on murine sleep, and suggest that melanopsin-based photoreception is primarily involved in sustaining light-induced sleep.


Assuntos
Luz , Opsinas de Bastonetes/fisiologia , Sono/efeitos da radiação , Animais , Escuridão , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fotoperíodo , Tempo de Reação , Opsinas de Bastonetes/genética , Sono/genética , Vigília
8.
PLoS Biol ; 7(6): e1000125, 2009 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-19513122

RESUMO

Light influences sleep and alertness either indirectly through a well-characterized circadian pathway or directly through yet poorly understood mechanisms. Melanopsin (Opn4) is a retinal photopigment crucial for conveying nonvisual light information to the brain. Through extensive characterization of sleep and the electrocorticogram (ECoG) in melanopsin-deficient (Opn4(-/-)) mice under various light-dark (LD) schedules, we assessed the role of melanopsin in mediating the effects of light on sleep and ECoG activity. In control mice, a light pulse given during the habitual dark period readily induced sleep, whereas a dark pulse given during the habitual light period induced waking with pronounced theta (7-10 Hz) and gamma (40-70 Hz) activity, the ECoG correlates of alertness. In contrast, light failed to induce sleep in Opn4(-/-) mice, and the dark-pulse-induced increase in theta and gamma activity was delayed. A 24-h recording under a LD 1-hratio1-h schedule revealed that the failure to respond to light in Opn4(-/-) mice was restricted to the subjective dark period. Light induced c-Fos immunoreactivity in the suprachiasmatic nuclei (SCN) and in sleep-active ventrolateral preoptic (VLPO) neurons was importantly reduced in Opn4(-/-) mice, implicating both sleep-regulatory structures in the melanopsin-mediated effects of light. In addition to these acute light effects, Opn4(-/-) mice slept 1 h less during the 12-h light period of a LD 12ratio12 schedule owing to a lengthening of waking bouts. Despite this reduction in sleep time, ECoG delta power, a marker of sleep need, was decreased in Opn4(-/-) mice for most of the (subjective) dark period. Delta power reached after a 6-h sleep deprivation was similarly reduced in Opn4(-/-) mice. In mice, melanopsin's contribution to the direct effects of light on sleep is limited to the dark or active period, suggesting that at this circadian phase, melanopsin compensates for circadian variations in the photo sensitivity of other light-encoding pathways such as rod and cones. Our study, furthermore, demonstrates that lack of melanopsin alters sleep homeostasis. These findings call for a reevaluation of the role of light on mammalian physiology and behavior.


Assuntos
Ritmo Circadiano/efeitos da radiação , Homeostase/efeitos da radiação , Luz , Opsinas de Bastonetes/deficiência , Opsinas de Bastonetes/metabolismo , Filtro Sensorial/efeitos da radiação , Sono/efeitos da radiação , Animais , Escuridão , Eletroencefalografia , Galanina/metabolismo , Camundongos , Neurônios/metabolismo , Neurônios/efeitos da radiação , Área Pré-Óptica/metabolismo , Área Pré-Óptica/efeitos da radiação , Proteínas Proto-Oncogênicas c-fos/metabolismo , Sono/fisiologia , Sono REM/efeitos da radiação , Núcleo Supraquiasmático/metabolismo , Núcleo Supraquiasmático/efeitos da radiação , Fatores de Tempo , Vigília/efeitos da radiação
9.
Sleep ; 45(4)2022 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-34618890

RESUMO

Down syndrome (DS) is a genetic disorder caused by the presence of all or part of the third copy of chromosome 21. DS is associated with cognitive disabilities, for which there are no drug therapies. In spite of significant behavioral and pharmacological efforts to treat cognitive disabilities, new and continued efforts are still necessary. Over 60% of children with DS are reported to have sleep apnea that disrupt normal sleep. Normal and adequate sleep is necessary to maintain optimal cognitive functions. Therefore, we asked whether improved quality and/or quantity of sleep could improve cognitive capacities of people with DS. To investigate this possibility, we used the Ts65Dn mouse model of DS and applied two methods for enhancing their sleep following training on mouse memory tasks. A behavioral method was to impose sleep deprivation prior to training resulting in sleep rebound following the training. A pharmacologic method, hypocretin receptor 2 antagonist, was used immediately after the training to enhance subsequent sleep knowing that hypocretin is involved in the maintenance of wake. Our behavioral method resulted in a sleep reorganization that decreased wake and increased rapid eye movement sleep following the training associated with an improvement of recognition memory and spatial memory in the DS model mice. Our pharmacologic approach decreased wake and increased non-rapid eye movement sleep and was associated with improvement only in the spatial memory task. These results show that enhancing sleep after the training in a memory task improves memory consolidation in a mouse model of DS.


Assuntos
Síndrome de Down , Animais , Cognição , Modelos Animais de Doenças , Síndrome de Down/complicações , Síndrome de Down/tratamento farmacológico , Síndrome de Down/genética , Humanos , Camundongos , Camundongos Transgênicos , Reconhecimento Psicológico , Sono
10.
Neurol Ther ; 11(3): 1353-1374, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35779189

RESUMO

INTRODUCTION: This randomized, double-blind, placebo-controlled study in healthy volunteers assessed the safety, tolerability, and pharmacokinetics of single ascending doses of intravenously administered NX210-a linear peptide derived from subcommissural organ-spondin-and explored the effects on blood/urine biomarkers and cerebral activity. METHODS: Participants in five cohorts (n = 8 each) were randomized to receive a single intravenous dose of NX210 (n = 6 each) (0.4, 1.25, 2.5, 5, and 10 mg/kg) or placebo (n = 2 each); in total, 10 and 29 participants received placebo and NX210, respectively. Blood samples were collected for pharmacokinetics within 180 min post dosing. Plasma and urine were collected from participants (cohorts: 2.5, 5, and 10 mg/kg) for biomarker analysis and electroencephalography (EEG) recordings within 48 h post dosing. Safety/tolerability and pharmacokinetic data were assessed before ascending to the next dose. RESULTS: The study included 39 participants. All dosages were safe and well tolerated. All treatment-emergent adverse events (n = 17) were of mild severity and resolved spontaneously (except one with unknown outcome). Twelve treatment-emergent adverse events (70.6%) were deemed drug related; seven of those (58.3%) concerned nervous system disorders (dizziness, headache, and somnolence). The pharmacokinetic analysis indicated a short half-life in plasma (6-20 min), high apparent volume of distribution (1870-4120 L), and rapid clearance (7440-16,400 L/h). In plasma, tryptophan and homocysteine showed dose-related increase and decrease, respectively. No drug dose effect was found for the glutamate or glutamine plasma biomarkers. Nevertheless, decreased blood glutamate and increased glutamine were observed in participants treated with NX210 versus placebo. EEG showed a statistically significant decrease in beta and gamma bands and a dose-dependent increasing trend in alpha bands. Pharmacodynamics effects were sustained for several hours (plasma) or 48 h (urine and EEG). CONCLUSION: NX210 is safe and well tolerated and may exert beneficial effects on the central nervous system, particularly in terms of cognitive processing.

11.
Clocks Sleep ; 2(4): 523-535, 2020 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-33271811

RESUMO

This study examined whether theta oscillations were compromised by the type of circadian disruption that impairs hippocampal-dependent memory processes. In prior studies on Siberian hamsters, we developed a one-time light treatment that eliminated circadian timing in the central pacemaker, the suprachiasmatic nucleus (SCN). These arrhythmic animals had impaired hippocampal-dependent memory whereas animals made arrhythmic with SCN lesions did not. The current study examined whether theta oscillations are compromised by the same light treatment that produced memory impairments in these animals. We found that both methods of inducing circadian-arrhythmia shortened theta episodes in the EEG by nearly 50%. SCN-lesioned animals, however, exhibited a 3-fold increase in the number of theta episodes and more than doubled the total time that theta dominated the EEG compared to SCN-intact circadian-arrhythmic animals. Video tracking showed that changes in theta were paralleled by similar changes in exploration behavior. These results suggest that the circadian-arrhythmic SCN interferes with hippocampal memory encoding by fragmenting theta oscillations. SCN-lesioned animals can, however, compensate for the shortened theta episodes by increasing their frequency. Implications for rhythm coherence and theta sequence models of memory formation are discussed.

12.
Artigo em Inglês | MEDLINE | ID: mdl-32195448

RESUMO

The Ts65Dn mouse is a well-studied model of trisomy 21, Down syndrome. This mouse strain has severe learning disability as measured by several rodent learning tests that depend on hippocampal spatial memory function. Hippocampal long-term potentiation (LTP) is deficient in these mice. Short-term daily treatment with low-dose GABA receptor antagonists rescue spatial learning and LTP in Ts65Dn mice leading to the hypothesis that the learning disability is due to GABAergic over-inhibition of hippocampal circuits. The fact that the GABA receptor antagonists were only effective if delivered during the daily light phase suggested that the source of the excess GABA was controlled directly or indirectly by the circadian system. The central circadian pacemaker of mammals is the suprachiasmatic nucleus (SCN), which is largely a GABAergic nucleus. In this study we investigated whether elimination of the SCN in Ts65Dn mice would restore their ability to form recognition memories as tested by the novel object recognition (NOR) task. Full, but not partial lesions of the SCN of Ts65Dn mice normalized their ability to perform on the NOR test. These results suggest that the circadian system modulates neuroplasticity over the time frame involved in the process of consolidation of recognition memories.

13.
Annu Int Conf IEEE Eng Med Biol Soc ; 2019: 6714-6717, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31947382

RESUMO

Polysomnography (PSG) is a multi-parametric test used in the study of sleep and as a diagnostic tool in sleep medicine. PSG is the gold standard that manually quantifies the apnea-hypopnea index (AHI) to assess the severity of sleep apnea syndrome (SAS). This work presents a novel method based on a dual tri-axis accelerometer system (Adaptive Accelerometry Derived Respiration, ADR) which was patched on the subject's chest that adaptively reconstructed thoracic and abdominal respiratory efforts. Performance evaluation was performed on a 60s-epoch basis using signal and physiological indicators: the evaluation consisted in the comparison of airflow estimations from ADR and RIP to the nasal airflow, considered as reference. Results showed that 74% of the 60s-epoch ADR airflow estimation present a correlation coefficient with nasal airflow ≥ 70% compared to 64% for RIP. Relative errors for one-minute respiration rate and tidal volume estimation appeared to be relatively low which reflected the good feasibility of the adaptive ADR method for respiration monitoring during sleep.


Assuntos
Respiração , Acelerometria , Humanos , Pletismografia , Polissonografia , Sono , Síndromes da Apneia do Sono
14.
Neurobiol Dis ; 30(1): 1-7, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18282758

RESUMO

Down syndrome is characterized by a host of behavioral abnormalities including sleep disturbances. Sleep and EEG was studied at the age of 3 months in two mouse models of the condition, Ts65Dn and Ts1Cje, carrying one extra copy of partially overlapping segments of the mmu chromosome 16 (equivalent to the human chromosome 21). We found that the Ts65Dn mice showed increased waking amounts at the expense of non-REM sleep, increased theta power during sleep and a delayed sleep rebound after sleep deprivation. In contrast, Ts1Cje had limited sleep and EEG abnormalities, showing only a delayed sleep rebound after sleep deprivation and no difference in theta power. We previously found that mice over-expressing the human APPwt transgene, a gene triplicated in Ts65Dn but not Ts1Cje, also show increased wake and theta power during sleep. These results demonstrate abnormalities in sleep and EEG in Ts65Dn mice and underscore a possible correlation between App overexpression and hippocampal theta oscillations.


Assuntos
Modelos Animais de Doenças , Síndrome de Down/genética , Síndrome de Down/fisiopatologia , Eletroencefalografia , Sono/fisiologia , Análise de Variância , Animais , Eletromiografia , Análise de Fourier , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Deleção de Sequência , Sono/genética , Privação do Sono/fisiopatologia , Trissomia
15.
BMC Neurosci ; 7: 81, 2006 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-17184520

RESUMO

BACKGROUND: Nitric oxide (NO) is a multifunctional molecule synthesized by three isozymes of the NO synthase (NOSs) acting as a messenger/modulator and/or a potential neurotoxin. In rodents, the role of NOSs in sleep processes and throughout aging is now well established. For example, sleep parameters are highly deteriorated in senescence accelerated-prone 8 (SAMP8) mice, a useful animal model to study aging or age-associated disorders, while the inducible form of NOS (iNOS) is down-regulated within the cortex and the sleep-structures of the brainstem. Evidence is now increasing for a role of iNOS and resulting oxidative stress but not for the constitutive expressed isozyme (nNOS). To better understand the role of nNOS in the behavioural impairments observed in SAMP8 versus SAMR1 (control) animals, we evaluated age-related variations occurring in the nNOS expression and activity and nitrites/nitrates (NOx-) levels, in three brain areas (n = 7 animals in each group). Calibrated reverse transcriptase (RT) and real-time polymerase chain reaction (PCR) and biochemical procedures were used. RESULTS: We found that the levels of nNOS mRNA decreased in the cortex and the hippocampus of 8- vs 2-month-old animals followed by an increase in 12-vs 8-month-old animals in both strains. In the brainstem, levels of nNOS mRNA decreased in an age-dependent manner in SAMP8, but not in SAMR1. Regional age-related changes were also observed in nNOS activity. Moreover, nNOS activity in hippocampus was found lower in 8-month-old SAMP8 than in SAMR1, while in the cortex and the brainstem, nNOS activities increased at 8 months and afterward decreased with age in SAMP8 and SAMR1. NOx- levels showed profiles similar to nNOS activities in the cortex and the brainstem but were undetectable in the hippocampus of SAMP8 and SAMR1. Finally, NOx- levels were higher in the cortex of 8 month-old SAMP8 than in age-matched SAMR1. CONCLUSION: Concomitant variations occurring in NO levels derived from nNOS and iNOS at an early age constitute a major factor of risk for sleep and/or memory impairments in SAMP8.


Assuntos
Envelhecimento/metabolismo , Encéfalo/metabolismo , Óxido Nítrico Sintase Tipo I/biossíntese , RNA Mensageiro/biossíntese , Fatores Etários , Envelhecimento/genética , Animais , Tronco Encefálico/metabolismo , Córtex Cerebral/metabolismo , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Mutantes , Óxido Nítrico Sintase Tipo I/genética , RNA Mensageiro/genética
16.
Neurobiol Aging ; 26(2): 265-73, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15582754

RESUMO

Changes occurring with age in cortical EEG and sleep-wake states architecture were examined in senescence accelerated prone (SAMP8) or senescence resistant (SAMR1) mice (age: 2 and 12 months) under baseline conditions or after a 4 h sleep deprivation (SD). In baseline conditions, an increase in slow wave sleep (SWS) amount (21-24%) occurs at the expense of the wakefulness (W) in old SAMP8 and SAMR1 mice versus young animals. In these conditions, SWS latency is reduced (67-72%). Moreover, in SAMP8 and SAMR1 mice, aging deteriorates paradoxical sleep (PS) architecture with more pronounced changes in SAMP8 (amount: -63%; episode duration: -44%; latency: +286%; circadian component loss; and EEG theta (theta) peak frequency (TPF): -1 Hz). During the 4 h recovery subsequent to a 4 h sleep deprivation, old SAMP8 mice exhibit an enhanced sensitivity resulting in SWS (+62%) and PS (+120%) rebounds, a characteristic of this inbred strain. Results obtained are discussed in line with the age-related learning and memory impairments existing in SAMP8 animals. In particular, the reduced cognitive performances described in old SAMP8 might be linked to the TPF deterioration during PS.


Assuntos
Envelhecimento/fisiologia , Eletroencefalografia , Sono/fisiologia , Vigília/fisiologia , Fatores Etários , Envelhecimento/genética , Animais , Comportamento Animal , Ritmo Circadiano/fisiologia , Demência/genética , Demência/fisiopatologia , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos , Transtornos do Sono do Ritmo Circadiano/fisiopatologia , Análise Espectral/métodos
17.
Neurobiol Aging ; 26(10): 1375-84, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16243608

RESUMO

Evidence that nitric oxide (NO) is involved in the regulation of rapid-eye-movement sleep (REMS) is supported by recent studies. During aging, NO generation encounters marked changes mainly related to the activation of the inducible NO-synthase (iNOS). To investigate links existing between iNOS and REMS impairments related to aging, we examine the age-related variations occurring in: mRNA and activity of iNOS in brainstem and frontal cortex; sleep parameters under baseline and after treatment by a selective iNOS inhibitor (AMT) in Senescence Accelerated Mice (SAM). SAMR1 (control) mice are a model of aging while SAMP8 are adequate to study neurodegenerative processes. RT-PCR analysis does not reveal significant variation in iNOS mRNA expression in both strains. However, significant age-related increases in iNOS activity occur in SAMR1 but such variation is not observed in SAMP8. In baseline conditions, aging induces a slight increase in slow-wave sleep (SWS) amounts in both groups and deteriorates greatly REMS architecture in SAMP8 compared to SAMR1. AMT reduces REMS amounts for 4-6h after treatment in a dose and age-dependent manner in SAMR1. Almost no changes occur in SAMP8. Data reported suggest that NO derived from iNOS contributes to trigger and maintain REMS during aging.


Assuntos
Envelhecimento/fisiologia , Encéfalo/metabolismo , Regulação Enzimológica da Expressão Gênica/fisiologia , Óxido Nítrico Sintase Tipo II/fisiologia , Sono REM/fisiologia , Fatores Etários , Envelhecimento/genética , Análise de Variância , Animais , Encéfalo/anatomia & histologia , Relação Dose-Resposta a Droga , Eletroencefalografia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Masculino , Camundongos , Camundongos Endogâmicos , Modelos Animais , Óxido Nítrico Sintase Tipo II/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Fases do Sono/genética , Fases do Sono/fisiologia , Cloreto de Sódio/farmacologia , Fatores de Tempo , Triazóis/farmacologia , Vigília/fisiologia
18.
Sleep Med Rev ; 9(2): 101-13, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15737789

RESUMO

Nitric oxide (NO) is a biological messenger synthesized by three main isoforms of NO synthase (NOS): neuronal (nNOS, constitutive calcium dependent), endothelial (eNOS, constitutive, calcium dependent) and inducible (iNOS, calcium independent). NOS is distributed in the brain either in circumscribed neuronal sets or in sparse interneurons. Within the laterodorsal tegmentum (LDT), pedunculopontine tegmentum and dorsal raphe nucleus, NOS-containing neurons overlap neurons grouped according to their contribution to sleep mechanisms. The main target for NO is the soluble guanylate cyclase that triggers an overproduction of cyclic guanosine monophosphate. NO in neurons of the pontine tegmentum facilitates sleep (particularly rapid-eye-movement sleep), and NO contained within the LDT intervenes in modulating the discharge of the neurons through an auto-inhibitory process involving the co-synthesized neurotransmitters. Moreover, NO synthesized within cholinergic neurons of the basal forebrain, while under control of the LDT, may modulate the spectral components of the EEG instead of the amounts of different sleep states. Finally, impairment of NO production (e.g. neurodegeneration, iNOS induction) has identifiable effects, including ageing, neuropathologies and parasitaemia.


Assuntos
Óxido Nítrico/fisiologia , Sono/fisiologia , Encéfalo/metabolismo , Guanilato Ciclase/metabolismo , Homeostase/fisiologia , Humanos , Neurônios/metabolismo , Óxido Nítrico/biossíntese , Sono REM/fisiologia
19.
PLoS One ; 10(6): e0128175, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26083020

RESUMO

Light has direct effects on sleep and wakefulness causing arousal in diurnal animals and sleep in nocturnal animals. In the present study, we assessed the modulation of light-induced sleep by melanopsin and the histaminergic system by exposing mice to millisecond light flashes and continuous light respectively. First, we show that the induction of sleep by millisecond light flashes is dose dependent as a function of light flash number. We found that exposure to 60 flashes of light occurring once every 60 seconds for 1-h (120-ms of total light over an hour) induced a similar amount of sleep as a continuous bright light pulse. Secondly, the induction of sleep by millisecond light flashes was attenuated in the absence of melanopsin when animals were presented with flashes occurring every 60 seconds over a 3-h period beginning at ZT13. Lastly, the acute administration of a histamine H3 autoreceptor antagonist, ciproxifan, blocked the induction of sleep by a 1-h continuous light pulse during the dark period. Ciproxifan caused a decrease in NREMS delta power and an increase in theta activity during both sleep and wake periods respectively. The data suggest that some form of temporal integration occurs in response to millisecond light flashes, and that this process requires melanopsin photoreception. Furthermore, the pharmacological data suggest that the increase of histaminergic neurotransmission is sufficient to attenuate the light-induced sleep response during the dark period.


Assuntos
Antagonistas dos Receptores Histamínicos/farmacologia , Imidazóis/farmacologia , Luz , Receptores Histamínicos H3/metabolismo , Opsinas de Bastonetes/metabolismo , Sono/efeitos dos fármacos , Animais , Eletroencefalografia , Eletromiografia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Receptores Histamínicos H3/química , Opsinas de Bastonetes/genética , Sono/efeitos da radiação , Vigília/efeitos dos fármacos , Vigília/efeitos da radiação
20.
Nat Commun ; 6: 8516, 2015 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-26465715

RESUMO

Many of the factors affecting the success of haematopoietic cell transplantation are still unknown. Here we show in mice that donor sleep deprivation reduces the ability of its haematopoietic stem cells (HSCs) to engraft and reconstitute the blood and bone marrow of an irradiated recipient by more than 50%. We demonstrate that sleep deprivation downregulates the expression of microRNA (miR)-19b, a negative regulator of the suppressor of cytokine signalling (SOCS) genes, which inhibit HSC migration and homing. Accordingly, HSCs from sleep-deprived mice have higher levels of SOCS genes expression, lower migration capacity in vitro and reduced homing to the bone marrow in vivo. Recovery of sleep after sleep deprivation restored the reconstitution potential of the HSCs. Taken together, this study provides insights into cellular and molecular mechanisms underlying the effects of sleep deprivation on HSCs, emphasizing the potentially critical role of donor sleep in the success of bone marrow transplantation.


Assuntos
Hormônio do Crescimento/sangue , Transplante de Células-Tronco Hematopoéticas , MicroRNAs/metabolismo , Privação do Sono/sangue , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Animais , Movimento Celular , Células-Tronco Hematopoéticas/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína 3 Supressora da Sinalização de Citocinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA