Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Nat Methods ; 19(7): 871-880, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35681062

RESUMO

Regulation of receptor tyrosine kinase (RTK) activity is necessary for studying cell signaling pathways in health and disease. We developed a generalized approach for engineering RTKs optically controlled with far-red light. We targeted the bacterial phytochrome DrBphP to the cell surface and allowed its light-induced conformational changes to be transmitted across the plasma membrane via transmembrane helices to intracellular RTK domains. Systematic optimization of these constructs has resulted in optically regulated epidermal growth factor receptor, HER2, TrkA, TrkB, FGFR1, IR1, cKIT and cMet, named eDrRTKs. eDrRTKs induced downstream signaling in mammalian cells in tens of seconds. The ability to activate eDrRTKs with far-red light enabled spectral multiplexing with fluorescent probes operating in a shorter spectral range, allowing for all-optical assays. We validated eDrTrkB performance in mice and found that minimally invasive stimulation in the neocortex with penetrating via skull far-red light-induced neural activity, early immediate gene expression and affected sleep patterns.


Assuntos
Fitocromo , Animais , Membrana Celular/metabolismo , Luz , Mamíferos , Camundongos , Transdução de Sinais
2.
Stroke ; 55(6): 1641-1649, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38572660

RESUMO

BACKGROUND: The current management of patients with stroke with intravenous thrombolysis and endovascular thrombectomy is effective only when it is timely performed on an appropriately selected but minor fraction of patients. The development of novel adjunctive therapy is highly desired to reduce morbidity and mortality with stroke. Since endothelial dysfunction is implicated in the pathogenesis of stroke and is featured with suppressed endothelial nitric oxide synthase (eNOS) with concomitant nitric oxide deficiency, restoring endothelial nitric oxide represents a promising approach to treating stroke injury. METHODS: This is a preclinical proof-of-concept study to determine the therapeutic effect of transcranial treatment with a low-power near-infrared laser in a mouse model of ischemic stroke. The laser treatment was performed before the middle cerebral artery occlusion with a filament. To determine the involvement of eNOS phosphorylation, unphosphorylatable eNOS S1176A knock-in mice were used. Each measurement was analyzed by a 2-way ANOVA to assess the effect of the treatment on cerebral blood flow with laser Doppler flowmetry, eNOS phosphorylation by immunoblot analysis, and stroke outcomes by infarct volumes and neurological deficits. RESULTS: Pretreatment with a 1064-nm laser at an irradiance of 50 mW/cm2 improved cerebral blood flow, eNOS phosphorylation, and stroke outcomes. CONCLUSIONS: Near-infrared II photobiomodulation could offer a noninvasive and low-risk adjunctive therapy for stroke injury. This new modality using a physical parameter merits further consideration to develop innovative therapies to prevent and treat a wide array of cardiovascular diseases.


Assuntos
Terapia com Luz de Baixa Intensidade , Óxido Nítrico Sintase Tipo III , Animais , Óxido Nítrico Sintase Tipo III/metabolismo , Camundongos , Fosforilação , Terapia com Luz de Baixa Intensidade/métodos , Masculino , Acidente Vascular Cerebral , Camundongos Endogâmicos C57BL , Infarto da Artéria Cerebral Média , Circulação Cerebrovascular/fisiologia , AVC Isquêmico/metabolismo , Modelos Animais de Doenças
3.
Brain Behav Immun ; 62: 137-150, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28109896

RESUMO

Both sleep loss and pathogens can enhance brain inflammation, sleep, and sleep intensity as indicated by electroencephalogram delta (δ) power. The pro-inflammatory cytokine interleukin-1 beta (IL-1ß) is increased in the cortex after sleep deprivation (SD) and in response to the Gram-negative bacterial cell-wall component lipopolysaccharide (LPS), although the exact mechanisms governing these effects are unknown. The nucleotide-binding domain and leucine-rich repeat protein-3 (NLRP3) inflammasome protein complex forms in response to changes in the local environment and, in turn, activates caspase-1 to convert IL-1ß into its active form. SD enhances the cortical expression of the somnogenic cytokine IL-1ß, although the underlying mechanism is, as yet, unidentified. Using NLRP3-gene knockout (KO) mice, we provide evidence that NLRP3 inflammasome activation is a crucial mechanism for the downstream pathway leading to increased IL-1ß-enhanced sleep. NLRP3 KO mice exhibited reduced non-rapid eye movement (NREM) sleep during the light period. We also found that sleep amount and intensity (δ activity) were drastically attenuated in NLRP3 KO mice following SD (homeostatic sleep response), as well as after LPS administration, although they were enhanced by central administration of IL-1ß. NLRP3, ASC, and IL1ß mRNA, IL-1ß protein, and caspase-1 activity were greater in the somatosensory cortex at the end of the wake-active period when sleep propensity was high and after SD in wild-type but not NLRP3 KO mice. Thus, our novel and converging findings suggest that the activation of the NLRP3 inflammasome can modulate sleep induced by both increased wakefulness and a bacterial component in the brain.


Assuntos
Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Privação do Sono/metabolismo , Sono/fisiologia , Animais , Inflamassomos/genética , Camundongos , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Polissonografia , Transdução de Sinais/fisiologia , Privação do Sono/genética , Vigília/fisiologia
4.
Proc Natl Acad Sci U S A ; 110(50): 20272-7, 2013 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-24191004

RESUMO

Although the neural circuitry underlying homeostatic sleep regulation is little understood, cortical neurons immunoreactive for neuronal nitric oxide synthase (nNOS) and the neurokinin-1 receptor (NK1) have been proposed to be involved in this physiological process. By systematically manipulating the durations of sleep deprivation and subsequent recovery sleep, we show that activation of cortical nNOS/NK1 neurons is directly related to non-rapid eye movement (NREM) sleep time, NREM bout duration, and EEG δ power during NREM sleep, an index of preexisting homeostatic sleep drive. Conversely, nNOS knockout mice show reduced NREM sleep time, shorter NREM bouts, and decreased power in the low δ range during NREM sleep, despite constitutively elevated sleep drive. Cortical NK1 neurons are still activated in response to sleep deprivation in these mice but, in the absence of nNOS, they are unable to up-regulate NREM δ power appropriately. These findings support the hypothesis that cortical nNOS/NK1 neurons translate homeostatic sleep drive into up-regulation of NREM δ power through an NO-dependent mechanism.


Assuntos
Ondas Encefálicas/fisiologia , Córtex Cerebral/fisiologia , Interneurônios/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Sono/fisiologia , Animais , Contagem de Células , Eletroencefalografia , Eletromiografia , Imuno-Histoquímica , Camundongos , Ratos , Ratos Sprague-Dawley
5.
Mol Neurodegener ; 18(1): 93, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-38041158

RESUMO

BACKGROUND: Alzheimer's disease (AD) patients exhibit memory disruptions and profound sleep disturbances, including disruption of deep non-rapid eye movement (NREM) sleep. Slow-wave activity (SWA) is a major restorative feature of NREM sleep and is important for memory consolidation. METHODS: We generated a mouse model where GABAergic interneurons could be targeted in the presence of APPswe/PS1dE9 (APP) amyloidosis, APP-GAD-Cre mice. An electroencephalography (EEG) / electromyography (EMG) telemetry system was used to monitor sleep disruptions in these animals. Optogenetic stimulation of GABAergic interneurons in the anterior cortex targeted with channelrhodopsin-2 (ChR2) allowed us to examine the role GABAergic interneurons play in sleep deficits. We also examined the effect of optogenetic stimulation on amyloid plaques, neuronal calcium as well as sleep-dependent memory consolidation. In addition, microglial morphological features and functions were assessed using confocal microscopy and flow cytometry. Finally, we performed sleep deprivation during optogenetic stimulation to investigate whether sleep restoration was necessary to slow AD progression. RESULTS: APP-GAD-Cre mice exhibited impairments in sleep architecture including decreased time spent in NREM sleep, decreased delta power, and increased sleep fragmentation compared to nontransgenic (NTG) NTG-GAD-Cre mice. Optogenetic stimulation of cortical GABAergic interneurons increased SWA and rescued sleep impairments in APP-GAD-Cre animals. Furthermore, it slowed AD progression by reducing amyloid deposition, normalizing neuronal calcium homeostasis, and improving memory function. These changes were accompanied by increased numbers and a morphological transformation of microglia, elevated phagocytic marker expression, and enhanced amyloid ß (Aß) phagocytic activity of microglia. Sleep was necessary for amelioration of pathophysiological phenotypes in APP-GAD-Cre mice. CONCLUSIONS: In summary, our study shows that optogenetic targeting of GABAergic interneurons rescues sleep, which then ameliorates neuropathological as well as behavioral deficits by increasing clearance of Aß by microglia in an AD mouse model.


Assuntos
Doença de Alzheimer , Humanos , Camundongos , Animais , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Microglia/metabolismo , Camundongos Transgênicos , Optogenética , Cálcio/metabolismo , Sono , Neurônios GABAérgicos/metabolismo , Modelos Animais de Doenças , Precursor de Proteína beta-Amiloide/genética
6.
J Neurosci ; 31(16): 6028-40, 2011 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-21508228

RESUMO

Cataplexy, a sudden unexpected muscle paralysis, is a debilitating symptom of the neurodegenerative sleep disorder, narcolepsy. During these attacks, the person is paralyzed, but fully conscious and aware of their surroundings. To identify potential neurons that might serve as surrogate orexin neurons to suppress such attacks, the gene for orexin (hypocretin), a peptide lost in most human narcoleptics, was delivered into the brains of the orexin-ataxin-3 transgenic mouse model of human narcolepsy. Three weeks after the recombinant adenoassociated virus (rAAV)-mediated orexin gene transfer, sleep-wake behavior was assessed. rAAV-orexin gene delivery into neurons of the zona incerta (ZI), or the lateral hypothalamus (LH) blocked cataplexy. Orexin gene transfer into the striatum or in the melanin-concentrating hormone neurons in the ZI or LH had no such effect, indicating site specificity. In transgenic mice lacking orexin neurons but given rAAV-orexin, detectable levels of orexin-A were evident in the CSF, indicating release of the peptide from the surrogate neurons. Retrograde tracer studies showed that the amygdala innervates the ZI consistent with evidence that strong emotions trigger cataplexy. In turn, the ZI projects to the locus ceruleus, indicating that the ZI is part of a circuit that stabilizes motor tone. Our results indicate that these neurons might also be recruited to block the muscle paralysis in narcolepsy.


Assuntos
Cataplexia/terapia , Terapia Genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Narcolepsia/terapia , Neurônios/metabolismo , Neuropeptídeos/genética , Subtálamo/metabolismo , Animais , Cataplexia/genética , Modelos Animais de Doenças , Eletroencefalografia , Eletromiografia , Técnicas de Transferência de Genes , Genótipo , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Camundongos Transgênicos , Narcolepsia/genética , Neuropeptídeos/metabolismo , Orexinas , Sono
7.
Brain Res Bull ; 187: 181-198, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35850189

RESUMO

Sleep abnormalities are widely reported in patients with Alzheimer's disease (AD) and are linked to cognitive impairments. Sleep abnormalities could be potential biomarkers to detect AD since they are often observed at the preclinical stage. Moreover, sleep could be a target for early intervention to prevent or slow AD progression. Thus, here we review changes in brain oscillations observed during sleep, their connection to AD pathophysiology and the role of specific brain circuits. Slow oscillations (0.1-1 Hz), sleep spindles (8-15 Hz) and their coupling during non-REM sleep are consistently reduced in studies of patients and in AD mouse models although the timing and magnitude of these alterations depends on the pathophysiological changes and the animal model studied. Changes in delta (1-4 Hz) activity are more variable. Animal studies suggest that hippocampal sharp-wave ripples (100-250 Hz) are also affected. Reductions in REM sleep amount and slower oscillations during REM are seen in patients but less consistently in animal models. Thus, changes in a variety of sleep oscillations could impact sleep-dependent memory consolidation or restorative functions of sleep. Recent mechanistic studies suggest that alterations in the activity of GABAergic neurons in the cortex, hippocampus and thalamic reticular nucleus mediate sleep oscillatory changes in AD and represent a potential target for intervention. Longitudinal studies of the timing of AD-related sleep abnormalities with respect to pathology and dysfunction of specific neural networks are needed to identify translationally relevant biomarkers and guide early intervention strategies to prevent or delay AD progression.


Assuntos
Doença de Alzheimer , Neurônios GABAérgicos , Animais , Eletroencefalografia , Neurônios GABAérgicos/fisiologia , Hipocampo/fisiologia , Camundongos , Sono/fisiologia , Tálamo/fisiologia
8.
MethodsX ; 9: 101811, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36065218

RESUMO

Studying the biology of sleep requires accurate and efficient assessment of the sleep stages. However, analysis of sleep-wake cycles in mice and other laboratory animals remains a time-consuming and laborious process. In this study, we developed a Python script and a process for the streamlined analysis of sleep data that includes real-time processing of electroencephalogram (EEG) and electromyogram (EMG) signals that is compatible with commercial sleep-recording software that supports user datagram protocol (UDP) communication. The process consists of EEG/EMG data acquisition, automated threshold calculation for real-time determination of sleep stages, sleep staging and EEG power spectrum analysis. It also allows data storage in the format that facilitates further analysis of the sleep pattern in mice. The described method is aimed at increasing efficiency of sleep stage scoring and analysis in mice thus facilitating sleep research. • A process of EEG/EMG recording and streamline analysis of sleep-wake cycle in real time in mice. • The compatibility with commercial sleep-recording software that can generate a UDP stream. • The capability of further analysis of recorded data by an open-source software.

9.
Sci Rep ; 12(1): 12278, 2022 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-35853986

RESUMO

Non-invasive stimulation technologies are emerging as potential treatment options for a range of neurodegenerative disorders. Experimental evidence suggests that stimuli-evoked changes in slow brain rhythms may mitigate or even prevent neuropathological and behavioral impairments. Slow wave activity is prevalent during sleep and can be triggered non-invasively by sensory stimulation targeting the visual system or directly via activation of neurons locally using optogenetics. Here, we developed new tools for delivering visual stimulation using light-emitting diodes in freely moving mice while awake and during sleep. We compared these tools to traditional optogenetic approaches used for local stimulation of neurons in the cerebral cortex. We then used these tools to compare the effects of low-frequency visual versus optogenetic stimulations on the slow wave activity and sleep pattern in mice. Visual stimulation effectively enhanced slow wave activity without disrupting the sleep pattern. Optogenetic stimulation of cortical GABAergic neurons increased NREM sleep. These results suggest that visual stimulation can be effective at boosting slow wave activity without having adverse effects on sleep and thus holds great potential as a non-invasive stimulation treatment strategy.


Assuntos
Sono de Ondas Lentas , Animais , Eletroencefalografia , Camundongos , Optogenética , Estimulação Luminosa , Sono/fisiologia , Sono de Ondas Lentas/fisiologia , Vigília/fisiologia
10.
Commun Biol ; 5(1): 1323, 2022 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-36460716

RESUMO

Alzheimer's disease (AD) is characterized by progressive memory loss and cognitive decline. These impairments correlate with early alterations in neuronal network activity in AD patients. Disruptions in the activity of individual neurons have been reported in mouse models of amyloidosis. However, the impact of amyloid pathology on the spontaneous activity of distinct neuronal types remains unexplored in vivo. Here we use in vivo calcium imaging with multiphoton microscopy to monitor and compare the activity of excitatory and two types of inhibitory interneurons in the cortices of APP/PS1 and control mice under isoflurane anesthesia. We also determine the relationship between amyloid accumulation and the deficits in spontaneous activity in APP/PS1 mice. We show that somatostatin-expressing (SOM) interneurons are hyperactive, while parvalbumin-expressing interneurons are hypoactive in APP/PS1 mice. Only SOM interneuron hyperactivity correlated with proximity to amyloid plaque. These inhibitory deficits were accompanied by decreased excitatory neuron activity in APP/PS1 mice. Our study identifies cell-specific neuronal firing deficits in APP/PS1 mice driven by amyloid pathology. These findings highlight the importance of addressing the complexity of neuron-specific deficits to ameliorate circuit dysfunction in Alzheimer's disease.


Assuntos
Doença de Alzheimer , Amiloidose , Camundongos , Animais , Interneurônios , Neurônios , Modelos Animais de Doenças , Placa Amiloide , Proteínas Amiloidogênicas
11.
Proc Natl Acad Sci U S A ; 105(29): 10227-32, 2008 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-18645184

RESUMO

The presence of large-amplitude, slow waves in the EEG is a primary characteristic that distinguishes cerebral activity during sleep from that which occurs during wakefulness. Although sleep-active neurons have been identified in other brain areas, neurons that are specifically activated during slow-wave sleep have not previously been described in the cerebral cortex. We have identified a population of cells in the cortex that is activated during sleep in three mammalian species. These cortical neurons are a subset of GABAergic interneurons that express neuronal NOS (nNOS). Because Fos expression in these sleep-active, nNOS-immunoreactive (nNOS-ir) neurons parallels changes in the intensity of slow-wave activity in the EEG, and these neurons are innvervated by neurotransmitter systems previously implicated in sleep/wake control, cortical nNOS-ir neurons may be part of the neurobiological substrate that underlies homeostatic sleep regulation.


Assuntos
Córtex Cerebral/citologia , Córtex Cerebral/fisiologia , Neurônios/classificação , Neurônios/fisiologia , Sono/fisiologia , Animais , Cricetinae , Eletroencefalografia , Eletrofisiologia , Interneurônios/classificação , Interneurônios/citologia , Interneurônios/fisiologia , Masculino , Mesocricetus , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/citologia , Óxido Nítrico Sintase Tipo I/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Privação do Sono/patologia , Privação do Sono/fisiopatologia , Vigília/fisiologia
12.
J Neurosci ; 29(12): 3705-14, 2009 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-19321767

RESUMO

Thyrotropin-releasing hormone (TRH) has previously been shown to promote wakefulness and to induce arousal from hibernation. Expression of TRH-R1 (TRH receptor 1) is enriched in the tuberal and lateral hypothalamic area (LHA), brain regions in which the hypocretin/orexin (Hcrt) cells are located. Because the Hcrt system is implicated in sleep/wake control, we hypothesized that TRH provides modulatory input to the Hcrt cells. In vitro electrophysiological studies showed that bath application of TRH caused concentration-dependent membrane depolarization, decreased input resistance, and increased firing rate of identified Hcrt neurons. In the presence of tetrodotoxin, TRH induced inward currents that were associated with a decrease in frequency, but not amplitude, of miniature postsynaptic currents (PSCs). Ion substitution experiments suggested that the TRH-induced inward current was mediated in part by Ca(2+) influx. Although TRH did not significantly alter either the frequency or amplitude of spontaneous excitatory PSCs, TRH (100 nm) increased the frequency of spontaneous inhibitory PSCs by twofold without affecting the amplitude of these events, indicating increased presynaptic GABA release onto Hcrt neurons. In contrast, TRH significantly reduced the frequency, but not amplitude, of miniature excitatory PSCs without affecting miniature inhibitory PSC frequency or amplitude, indicating that TRH also reduces the probability of glutamate release onto Hcrt neurons. When injected into the LHA, TRH increased locomotor activity in wild-type mice but not in orexin/ataxin-3 mice in which the Hcrt neurons degenerate postnatally. Together, these results are consistent with the hypothesis that TRH modulates behavioral arousal, in part, through the Hcrt system.


Assuntos
Nível de Alerta , Comportamento Animal , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Hormônio Liberador de Tireotropina/fisiologia , Potenciais de Ação , Animais , Temperatura Corporal , Cálcio/metabolismo , Feminino , Região Hipotalâmica Lateral/citologia , Região Hipotalâmica Lateral/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Camundongos , Camundongos Transgênicos , Potenciais Pós-Sinápticos em Miniatura , Atividade Motora , Neuropeptídeos/genética , Orexinas , Técnicas de Patch-Clamp , Hormônio Liberador de Tireotropina/farmacologia
13.
Front Neurosci ; 14: 705, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32714142

RESUMO

Alzheimer's disease (AD) is the major cause of dementia, characterized by the presence of amyloid-beta plaques and neurofibrillary tau tangles. Plaques and tangles are associated with sleep-wake cycle disruptions, including the disruptions in non-rapid eye movement (NREM) slow wave sleep (SWS). Alzheimer's patients spend less time in NREM sleep and exhibit decreased slow wave activity (SWA). Consistent with the critical role of SWS in memory consolidation, reduced SWA is associated with impaired memory consolidation in AD patients. The aberrant SWA can be modeled in transgenic mouse models of amyloidosis and tauopathy. Animal models exhibited slow wave impairments early in the disease progression, prior to the deposition of amyloid-beta plaques, however, in the presence of abundant oligomeric amyloid-beta. Optogenetic rescue of SWA successfully halted the amyloid accumulation and restored intraneuronal calcium levels in mice. On the other hand, optogenetic acceleration of slow wave frequency exacerbated amyloid deposition and disrupted neuronal calcium homeostasis. In this review, we summarize the evidence and the mechanisms underlying the existence of a positive feedback loop between amyloid/tau pathology and SWA disruptions that lead to further accumulations of amyloid and tau in AD. Moreover, since SWA disruptions occur prior to the plaque deposition, SWA disruptions may provide an early biomarker for AD. Finally, we propose that therapeutic targeting of SWA in AD might lead to an effective treatment for Alzheimer's patients.

14.
J Neurosci ; 28(28): 7193-201, 2008 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-18614689

RESUMO

Sleep deprivation (SD) results in increased electroencephalographic (EEG) delta power during subsequent non-rapid eye movement sleep (NREMS) and is associated with changes in the expression of circadian clock-related genes in the cerebral cortex. The increase of NREMS delta power as a function of previous wake duration varies among inbred mouse strains. We sought to determine whether SD-dependent changes in circadian clock gene expression parallel this strain difference described previously at the EEG level. The effects of enforced wakefulness of incremental durations of up to 6 h on the expression of circadian clock genes (bmal1, clock, cry1, cry2, csnk1epsilon, npas2, per1, and per2) were assessed in AKR/J, C57BL/6J, and DBA/2J mice, three strains that exhibit distinct EEG responses to SD. Cortical expression of clock genes subsequent to SD was proportional to the increase in delta power that occurs in inbred strains: the strain that exhibits the most robust EEG response to SD (AKR/J) exhibited dramatic increases in expression of bmal1, clock, cry2, csnkIepsilon, and npas2, whereas the strain with the least robust response to SD (DBA/2) exhibited either no change or a decrease in expression of these genes and cry1. The effect of SD on circadian clock gene expression was maintained in mice in which both of the cryptochrome genes were genetically inactivated. cry1 and cry2 appear to be redundant in sleep regulation as elimination of either of these genes did not result in a significant deficit in sleep homeostasis. These data demonstrate transcriptional regulatory correlates to previously described strain differences at the EEG level and raise the possibility that genetic differences underlying circadian clock gene expression may drive the EEG differences among these strains.


Assuntos
Ritmo alfa , Córtex Cerebral/fisiologia , Ritmo Circadiano/genética , Regulação da Expressão Gênica/fisiologia , Privação do Sono/metabolismo , Análise de Variância , Animais , Proteínas CLOCK , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Criptocromos , Flavoproteínas/genética , Flavoproteínas/metabolismo , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Circadianas Period , Especificidade da Espécie , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
15.
Sleep ; 42(10)2019 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-31328777

RESUMO

Slow-wave activity (SWA) is an oscillatory neocortical activity occurring in the electroencephalogram delta (δ) frequency range (~0.5-4 Hz) during nonrapid eye movement sleep. SWA is a reliable indicator of sleep homeostasis after acute sleep loss and is involved in memory processes. Evidence suggests that cortical neuronal nitric oxide synthase (nNOS) expressing neurons that coexpress somatostatin (SST) play a key role in regulating SWA. However, previous studies lacked selectivity in targeting specific types of neurons that coexpress nNOS-cells which are activated in the cortex after sleep loss. We produced a mouse model that knocks out nNOS expression in neurons that coexpress SST throughout the cortex. Mice lacking nNOS expression in SST positive neurons exhibited significant impairments in both homeostatic low-δ frequency range SWA production and a recognition memory task that relies on cortical input. These results highlight that SST+/nNOS+ neurons are involved in the SWA homeostatic response and cortex-dependent recognition memory.


Assuntos
Córtex Cerebral/metabolismo , Ritmo Delta/fisiologia , Memória/fisiologia , Óxido Nítrico Sintase Tipo I/deficiência , Reconhecimento Psicológico/fisiologia , Somatostatina/deficiência , Animais , Eletroencefalografia/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neurônios/metabolismo , Óxido Nítrico Sintase Tipo I/genética , Sono/fisiologia , Somatostatina/genética
16.
J Neurosci ; 27(51): 14041-8, 2007 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-18094243

RESUMO

The hypocretin (HCRT) neurons are located only in the perifornical area of the lateral hypothalamus and heavily innervate the cholinergic neurons in the basal forebrain (BF), histamine neurons in the tuberomammillary nucleus (TMN), and the noradrenergic locus ceruleus (LC) neurons, three neuronal populations that have traditionally been implicated in regulating arousal. Based on the innervation, HCRT neurons may regulate arousal by driving these downstream arousal neurons. Here, we directly test this hypothesis by a simultaneous triple lesion of these neurons using three saporin-conjugated neurotoxins. Three weeks after lesion, the daily levels of wake were not changed in rats with double or triple lesions, although rats with triple lesions were asleep more during the light-to-dark transition period. The double- and triple-lesioned rats also had more stable sleep architecture compared with nonlesioned saline rats. These results suggest that the cholinergic BF, TMN, and LC neurons jointly modulate arousal at a specific circadian time, but they are not essential links in the circuitry responsible for daily levels of wake, as traditionally hypothesized.


Assuntos
Nível de Alerta/fisiologia , Neurônios/fisiologia , Periodicidade , Proteínas de Plantas/toxicidade , Proteínas Inativadoras de Ribossomos Tipo 1/toxicidade , Sono/fisiologia , Animais , Nível de Alerta/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Masculino , Neurônios/efeitos dos fármacos , Neuropeptídeos/fisiologia , Orexinas , Ratos , Ratos Sprague-Dawley , Saporinas , Sono/efeitos dos fármacos , Vigília/efeitos dos fármacos , Vigília/fisiologia
17.
J Neurochem ; 105(4): 1480-98, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18331290

RESUMO

Previous studies have supported the hypothesis that macromolecular synthesis occurs in the brain during sleep as a response to prior waking activities and that prostaglandin D2 (PGD2) is an endogenous sleep substance whose effects are dependent on adenosine A2a receptor-mediated signaling. We compared gene expression in the cerebral cortex, basal forebrain, and hypothalamus during PGD2-induced and adenosinergically-induced sleep to results from our previously published study of recovery sleep (RS) after sleep deprivation (SD). Immediate early gene expression in the cortex during sleep induced by PGD2- or by the selective adenosine A2a agonist CGS21680 showed limited similarity to that observed during RS while, in the basal forebrain and hypothalamus, widespread activation of immediate early genes not seen during RS occurred. In all three brain regions, PGD2 and CGS21680 reduced the expression of arc, a transcript whose expression is elevated during SD. Using GeneChips, the majority of genes induced by either PGD2 or CGS21680 were induced by both, suggesting activation of the same pathways. However, gene expression induced in the brain after PGD2 or CGS21680 treatment was distinct from that described during RS after SD and apparently involves glial cell gene activation and signaling pathways in neural-immune interactions.


Assuntos
Adenosina/fisiologia , Química Encefálica , Regulação da Expressão Gênica/fisiologia , Prostaglandina D2/fisiologia , Sono/fisiologia , Adenosina/análogos & derivados , Adenosina/farmacologia , Agonistas do Receptor A2 de Adenosina , Animais , Química Encefálica/efeitos dos fármacos , Química Encefálica/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Fenetilaminas/farmacologia , Ratos , Ratos Wistar , Receptor A2A de Adenosina/fisiologia , Sono/efeitos dos fármacos , Ativação Transcricional
18.
Neuroscience ; 379: 189-201, 2018 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-29438803

RESUMO

Slow-wave activity (SWA) in the electroencephalogram during slow-wave sleep (SWS) varies as a function of sleep-wake history. A putative sleep-active population of neuronal nitric oxide synthase (nNOS)-containing interneurons in the cerebral cortex, defined as such by the expression of Fos in animals euthanized after protracted deep sleep, may be a local regulator of SWA. We investigated whether electrophysiological responses to activation of these cells are consistent with their role of a local regulator of SWA. Using a Cre/loxP strategy, we targeted the population of nNOS interneurons to express the light-activated cation channel Channelrhodopsin2 and the histological marker tdTomato in mice. We then performed histochemical and optogenetic studies in these transgenic mice. Our studies provided histochemical evidence of transgene expression and electrophysiological evidence that the cerebral cortex was responsive to optogenetic manipulation of these cells in both anesthetized and behaving mice. Optogenetic stimulation of the cerebral cortex of animals expressing Channelrhodopsin2 in nNOS interneurons triggered an acute positive deflection of the local field potential that was followed by protracted oscillatory events only during quiet wake and slow wave sleep. The response during wake was maximal when the electroencephalogram (EEG) was in a negative polarization state and abolished when the EEG was in a positive polarization state. Since the polarization state of the EEG is a manifestation of slow-wave oscillations in the activity of underlying pyramidal neurons between the depolarized (LFP negative) and hyperpolarized (LFP positive) states, these data indicate that sleep-active cortical neurons expressing nNOS function in sleep slow-wave physiology.


Assuntos
Córtex Cerebral/fisiologia , Neurônios/fisiologia , Óxido Nítrico Sintase Tipo I/metabolismo , Sono de Ondas Lentas/fisiologia , Animais , Córtex Cerebral/citologia , Córtex Cerebral/fisiopatologia , Channelrhodopsins/genética , Channelrhodopsins/metabolismo , Eletrocorticografia , Eletromiografia , Potenciais Evocados , Masculino , Camundongos Transgênicos , Neurônios/citologia , Optogenética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Privação do Sono/fisiopatologia
19.
J Neurosci ; 26(31): 8092-100, 2006 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-16885223

RESUMO

It is currently hypothesized that the drive to sleep is determined by the activity of the basal forebrain (BF) cholinergic neurons, which release adenosine (AD), perhaps because of increased metabolic activity associated with the neuronal discharge during waking, and the accumulating AD begins to inhibit these neurons so that sleep-active neurons can become active. This hypothesis grew from the observation that AD induces sleep and AD levels increase with wake in the basal forebrain, but surprisingly it still remains untested. Here we directly test whether the basal forebrain cholinergic neurons are central to the AD regulation of sleep drive by administering 192-IgG-saporin to lesion the BF cholinergic neurons and then measuring AD levels in the BF. In rats with 95% lesion of the BF cholinergic neurons, AD levels in the BF did not increase with 6 h of prolonged waking. However, the lesioned rats had intact sleep drive after 6 and 12 h of prolonged waking, indicating that the AD accumulation in the BF is not necessary for sleep drive. Next we determined that, in the absence of the BF cholinergic neurons, the selective adenosine A1 receptor agonist N6-cyclohexyladenosine, administered to the BF, continued to be effective in inducing sleep, indicating that the BF cholinergic neurons are not essential to sleep induction. Thus, neither the activity of the BF cholinergic neurons nor the accumulation of AD in the BF during wake is necessary for sleep drive.


Assuntos
Potenciais de Ação/fisiologia , Adenosina/metabolismo , Homeostase/fisiologia , Neurônios/fisiologia , Prosencéfalo/fisiologia , Receptores Purinérgicos P1/metabolismo , Sono/fisiologia , Animais , Eletroencefalografia , Ratos , Ratos Sprague-Dawley
20.
Neurosci Lett ; 659: 44-47, 2017 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-28866052

RESUMO

Evidence indicates that the neuropeptide substance P (SP) can act through neurokinin receptors to alter sleep and/or non-rapid eye movement (NREM) sleep slow-wave activity. Consequently, drugs acting on SP receptors could potentially be used as a novel treatment for sleep-related disorders. In the present study, we used SP conjugated with cholera toxin A subunit (SP-CTA), which enhances its duration of activity on SP receptor-expressing cells, to determine the effects of selectively activating SP receptor-expressing brain cells on sleep regulation in mice. Herein, we found that intracerebroventricular administration of SP-CTA enhanced amounts of NREM sleep which was highly fragmented. This result suggests that the activation of SP receptor-expressing cells in the brain can produce not only arousal effects as shown in previous studies but also sleep-inducing effects.


Assuntos
Toxina da Cólera/farmacologia , Sono/efeitos dos fármacos , Substância P/farmacologia , Animais , Toxina da Cólera/administração & dosagem , Imunotoxinas/farmacologia , Infusões Intraventriculares , Masculino , Camundongos , Receptores da Neurocinina-1/metabolismo , Substância P/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA