Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Neurosci ; 16: 969712, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36340773

RESUMO

Non-rapid eye movement (NREM) sleep is tightly homeostatically regulated and essential for survival. In the electroencephalogram (EEG), oscillations in the delta (0.5-4 Hz) range are prominent during NREM sleep. These delta oscillations are, to date, the best indicator for homeostatic sleep regulation; they are increased after prolonged waking and fade during NREM sleep. The precise mechanisms underlying sleep homeostasis and the generation of EEG delta oscillations are still being investigated. Activity-dependent neuronal calcium influx has been hypothesized to play an important role in generating delta oscillations and might be involved in downstream signaling that mediates sleep function. Dihydropyridine blockers of L-type voltage-gated calcium channels (VGCCs) are in wide clinical use to treat hypertension and other cardiovascular disorders and are readily blood-brain-barrier penetrant. We therefore, wanted to investigate their potential effects on EEG delta oscillation and homeostatic NREM sleep regulation in freely behaving mice. In vivo two-photon imaging of cortical neurons showed larger spontaneous calcium transients in NREM sleep compared to waking. Application of the dihydropyridine calcium blocker nicardipine significantly reduced cortical calcium transients without affecting the generation of delta oscillations. Nicardipine also did not affect EEG delta oscillations over 24 h following application. The time spent in NREM sleep and NREM episode duration was also not affected. Thus, acute block of calcium entry through L-type VGCCs does not interfere with EEG delta oscillations or their homeostatic regulation, despite prior evidence from calcium channel knockout mice.

2.
Front Neurosci ; 16: 868049, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35812231

RESUMO

Acute cocaine disturbs sleep on a dose-dependent basis; however, the consequences of chronic cocaine remain unclear. While the arousal promotion following cocaine has been well-established, effects of cocaine on sleep after termination of chronic cocaine exposure appear variable in human subjects with few studies in non-human subjects. Here, a within-subjects design (outcomes normalized to baseline, undisturbed behavior) and between-subjects design (repeated experimenter-administered cocaine vs. experimenter-administered saline) was used to investigate sleep homeostasis and sleep/waking under repeated cocaine/saline exposure and prolonged forced abstinence conditions in mice. Overall, during the forced abstinence period increases in arousal, as determined by sleep latency and gamma energy, persisted for 2 weeks. However, the sleep response to externally enforced sleep deprivation was unchanged suggesting that sleep disruptions during the forced abstinence period were driven by enhancement of arousal in the absence of changes in sleep homeostatic responses.

3.
Pharmacol Biochem Behav ; 206: 173194, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33940055

RESUMO

Dopamine, orexin (hypocretin), and adenosine systems have dual roles in reward and sleep/arousal suggesting possible mechanisms whereby drugs of abuse may influence both reward and sleep/arousal. While considerable variability exists across studies, drugs of abuse such as cocaine induce an acute sleep loss followed by an immediate recovery pattern that is consistent with a normal response to loss of sleep. Under more chronic cocaine exposure conditions, an abnormal recovery pattern is expressed that includes a retention of sleep disturbance under withdrawal and into abstinence conditions. Conversely, experimentally induced sleep disturbance can increase cocaine seeking. Thus, complementary, sleep-related therapeutic approaches may deserve further consideration along with development of non-human models to better characterize sleep disturbance-reward seeking interactions across drug experience.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/psicologia , Cocaína/farmacologia , Transtornos do Sono-Vigília/psicologia , Sono/efeitos dos fármacos , Adenosina/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Cocaína/efeitos adversos , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Condicionamento Operante/efeitos dos fármacos , Dopamina/metabolismo , Inibidores da Captação de Dopamina/farmacologia , Feminino , Humanos , Masculino , Orexinas/metabolismo , Recompensa , Autoadministração , Transtornos do Sono-Vigília/metabolismo
4.
Neurosci Lett ; 740: 135450, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33127445

RESUMO

The rodent dorsal hippocampus is essential for episodic memory consolidation, a process heavily modulated by dopamine D1-like receptor (D1/5R) activation. It was previously thought that the ventral tegmental area provided the only supply of dopamine release to dorsal hippocampus, but several recent studies have established the locus coeruleus (LC) as the major source for CA1. Here we show that selective blockade of the norepinephrine transporter (NET) prevents dopamine-dependent, late long-term synaptic potentiation (LTP) in dorsal CA1, a neural correlate of memory formation that relies on LC-mediated activation of D1/5Rs. Since dopamine activation of D1/5Rs by vesicular release is expected to be enhanced by NET antagonism, our data identify NET reversal as a plausible mechanism for LC-mediated DA release. We also show that genetic deletion of LC NMDA receptors (NMDARs) blocks D1R-mediated LTP, suggesting the requirement of both a functional NET and presynaptic NMDARs for this release. As LC activity is highly correlated with attentional processes and memory, these experiments provide insight into how selective attention influences memory formation at the synaptic and circuit levels.


Assuntos
Dopamina/farmacologia , Hipocampo/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-ets/antagonistas & inibidores , Sinapses/efeitos dos fármacos , Antagonistas de Receptores Adrenérgicos alfa 2/farmacologia , Animais , Atenção/efeitos dos fármacos , Região CA1 Hipocampal/efeitos dos fármacos , Dopamina/metabolismo , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de N-Metil-D-Aspartato/genética
5.
eNeuro ; 7(6)2020.
Artigo em Inglês | MEDLINE | ID: mdl-33139319

RESUMO

Drug addiction and withdrawal are characterized by sleep disruption, but the effects of sleep disruption on these states are not well characterized. Sleep deprivation (SD) immediately before the cocaine conditioning trials enhanced cocaine conditioned place preference (CPP) in a dose-dependent manner (3, 8 mg/kg but not 15 mg/kg) in mice. SD immediately before the postconditioning test also enhanced cocaine CPP preference in a dose-dependent manner (8 mg/kg, but not 3, 15 mg/kg). Exposure to orexin-receptor antagonism (1 mg/kg SB 334867, an orexin 1 receptor antagonist; OX1R) just before cocaine-conditioning trials or the postconditioning test attenuated SD-enhanced preference. This suggests a potential therapeutic role for the manipulation of the orexin system to mitigate drug seeking, especially in the context of sleep loss before drug exposure.


Assuntos
Cocaína , Animais , Condicionamento Clássico , Camundongos , Antagonistas dos Receptores de Orexina/farmacologia , Receptores de Orexina , Privação do Sono
6.
J Neurosci ; 40(48): 9306-9316, 2020 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-33097636

RESUMO

Patients with sleeping sickness, caused by the parasite Trypanosoma brucei, have disruptions in both sleep timing and sleep architecture. However, the underlying cause of these sleep disturbances is not well understood. Here, we assessed the sleep architecture of male mice infected with T. brucei and found that infected mice had drastically altered sleep patterns. Interestingly, T. brucei-infected mice also had a reduced homeostatic sleep response to sleep deprivation, a response modulated by the adenosine system. We found that infected mice had a reduced electrophysiological response to an adenosine receptor antagonist and increased adenosine receptor gene expression. Although the mechanism by which T. brucei infection causes these changes remains to be determined, our findings suggest that the symptoms of sleeping sickness may be because of alterations in homeostatic adenosine signaling.SIGNIFICANCE STATEMENT Sleeping sickness is a fatal disease that disrupts the circadian clock, causes disordered temperature regulation, and induces sleep disturbance. To examine the neurologic effects of infection in the absence of other symptoms, in this study, we used a mouse model of sleeping sickness in which the acute infection was treated but brain infection remained. Using this model, we evaluated the effects of the sleeping sickness parasite, Trypanosoma brucei, on sleep patterns in mice, under both normal and sleep-deprived conditions. Our findings suggest that signaling of adenosine, a neuromodulator involved in mediating homeostatic sleep drive, may be reduced in infected mice.


Assuntos
Adenosina/fisiologia , Sono , Tripanossomíase Africana/fisiopatologia , Antagonistas do Receptor A2 de Adenosina/farmacologia , Animais , Eletroencefalografia , Eletromiografia , Fenômenos Eletrofisiológicos , Expressão Gênica , Homeostase , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptor A2A de Adenosina/genética , Receptor A2A de Adenosina/metabolismo , Privação do Sono , Trypanosoma brucei brucei
7.
Elife ; 92020 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-32851972

RESUMO

Neuronal activity and gene expression in response to the loss of sleep can provide a window into the enigma of sleep function. Sleep loss is associated with brain differential gene expression, an increase in pyramidal cell mEPSC frequency and amplitude, and a characteristic rebound and resolution of slow wave sleep-slow wave activity (SWS-SWA). However, the molecular mechanism(s) mediating the sleep-loss response are not well understood. We show that sleep-loss regulates MEF2C phosphorylation, a key mechanism regulating MEF2C transcriptional activity, and that MEF2C function in postnatal excitatory forebrain neurons is required for the biological events in response to sleep loss in C57BL/6J mice. These include altered gene expression, the increase and recovery of synaptic strength, and the rebound and resolution of SWS-SWA, which implicate MEF2C as an essential regulator of sleep function.


Assuntos
Córtex Cerebral/fisiologia , Regulação da Expressão Gênica , Privação do Sono , Animais , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Sono/fisiologia , Transcrição Gênica
8.
PLoS One ; 15(5): e0233561, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32470016

RESUMO

Cortical neurons fire intermittently and synchronously during non-rapid eye movement sleep (NREMS), in which active and silent periods are referred to as ON and OFF periods, respectively. Neuronal firing rates during ON periods (NREMS-ON-activity) are similar to those of wakefulness (W-activity), raising the possibility that NREMS-ON neuronal-activity is fragmented W-activity. To test this, we investigated the patterning and organization of cortical spike trains and of spike ensembles in neuronal networks using extracellular recordings in mice. Firing rates of neurons during NREMS-ON and W were similar, but showed enhanced bursting in NREMS with no apparent preference in occurrence, relative to the beginning or end of the on-state. Additionally, there was an overall increase in the randomness of occurrence of sequences comprised of multi-neuron ensembles in NREMS recorded from tetrodes. In association with increased burst firing, somatic calcium transients were increased in NREMS. The increased calcium transients associated with bursting during NREM may activate calcium-dependent, cell-signaling pathways for sleep related cellular processes.


Assuntos
Neurônios/fisiologia , Sono de Ondas Lentas , Vigília , Animais , Eletroencefalografia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
9.
Proc Natl Acad Sci U S A ; 117(19): 10547-10553, 2020 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-32350140

RESUMO

The activity-regulated cytoskeleton-associated protein (Arc) gene is a neural immediate early gene that is involved in synaptic downscaling and is robustly induced by prolonged wakefulness in rodent brains. Converging evidence has led to the hypothesis that wakefulness potentiates, and sleep reduces, synaptic strengthening. This suggests a potential role for Arc in these and other sleep-related processes. However, the role of Arc in sleep remains unknown. Here, we demonstrated that Arc is important for the induction of multiple behavioral and molecular responses associated with sleep homeostasis. Arc knockout (KO) mice displayed increased time spent in rapid eye movement (REM) sleep under baseline conditions and marked attenuation of sleep rebound to both 4 h of total sleep deprivation (SD) and selective REM deprivation. At the molecular level, the following homeostatic sleep responses to 4-h SD were all blunted in Arc KO mice: increase of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor GluA1 and its phosphorylation in synaptoneurosomes; induction of a subset of SD-response genes; and suppression of the GluA1 messenger RNA in the cortex. In wild-type brains, SD increased Arc protein expression in multiple subcellular locations, including the nucleus, cytoplasm, and synapse, which is reversed in part by recovery sleep. Arc is critical for these behavioral and multiple molecular responses to SD, thus providing a multifunctional role for Arc in the maintenance of sleep homeostasis, which may be attributed by the sleep/wake-associated changes in subcellular location of Arc.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Sono/fisiologia , Animais , Encéfalo/fisiologia , Núcleo Celular/metabolismo , Córtex Cerebral/fisiologia , Citoplasma/metabolismo , Proteínas do Citoesqueleto/genética , Eletroencefalografia/métodos , Homeostase/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Receptores de AMPA/metabolismo , Sono/genética , Privação do Sono/fisiopatologia , Sono REM/fisiologia , Vigília/genética , Vigília/fisiologia
10.
Sci Rep ; 10(1): 2278, 2020 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-32042079

RESUMO

Cortical networks exhibit large shifts in spontaneous dynamics depending on the vigilance state. Waking and rapid eye movement (REM) sleep are characterized by ongoing irregular activity of cortical neurons while during slow wave sleep (SWS) these neurons show synchronous alterations between silent (OFF) and active (ON) periods. The network dynamics underlying these phenomena are not fully understood. Additional information about the state of cortical networks can be obtained by evaluating evoked cortical responses during the sleep-wake cycle. We measured local field potentials (LFP) and multi-unit activity (MUA) in the cortex in response to repeated brief optogenetic stimulation of thalamocortical afferents. Both LFP and MUA responses were considerably increased in sleep compared to waking, with larger responses during SWS than during REM sleep. The strongly increased cortical response in SWS is discussed within the context of SWS-associated neuro-modulatory tone that may reduce feedforward inhibition. Responses to stimuli were larger during SWS-OFF periods than during SWS-ON periods. SWS responses showed clear daily fluctuation correlated to light-dark cycle, but no reaction to increased sleep need following sleep deprivation. Potential homeostatic synaptic plasticity was either absent or masked by large vigilance-state effects.


Assuntos
Córtex Cerebral/fisiologia , Sono REM/fisiologia , Sono de Ondas Lentas/fisiologia , Tálamo/fisiologia , Vigília/fisiologia , Animais , Córtex Cerebral/citologia , Eletroencefalografia , Masculino , Camundongos , Modelos Animais , Inibição Neural/fisiologia , Vias Neurais/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Optogenética , Fotoperíodo , Tálamo/citologia
11.
Front Neurosci ; 13: 740, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31379490

RESUMO

Roughly one-third of the human lifetime is spent in sleep, yet the reason for sleep remains unclear. Understanding the physiologic function of sleep is crucial toward establishing optimal health. Several proposed concepts address different aspects of sleep physiology, including humoral and circuit-based theories of sleep-wake regulation, the homeostatic two-process model of sleep regulation, the theory of sleep as a state of adaptive inactivity, and observations that arousal state and sleep homeostasis can be dissociated in pathologic disorders. Currently, there is no model that places the regulation of arousal and sleep homeostasis in a unified conceptual framework. Adenosine is well known as a somnogenic substance that affects normal sleep-wake patterns through several mechanisms in various brain locations via A1 or A2A receptors (A1Rs or A2ARs). Many cells and processes appear to play a role in modulating the extracellular concentration of adenosine at neuronal A1R or A2AR sites. Emerging evidence suggests that A1Rs and A2ARs have different roles in the regulation of sleep. In this review, we propose a model in which A2ARs allow the brain to sleep, i.e., these receptors provide sleep gating, whereas A1Rs modulate the function of sleep, i.e., these receptors are essential for the expression and resolution of sleep need. In this model, sleep is considered a brain state established in the absence of arousing inputs.

12.
Sci Rep ; 8(1): 17672, 2018 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-30518935

RESUMO

Repeated exposure to drugs of abuse progressively increases the response to the same stimuli, a process known as sensitization. Behavioral sensitization to cocaine administration is often measured in non-human subjects via locomotor activity which is easily quantifiable. The effects of four hours of sleep deprivation on repeated cocaine (five daily and one challenge) showed attenuated hyperactivity on the first day only, compared to the non-deprived group. Both groups reached the same final level of sensitization, indicating that sleep deprivation altered the time course, but not magnitude of locomotor sensitization.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/complicações , Cocaína/farmacologia , Inibidores da Captação de Dopamina/farmacologia , Locomoção/efeitos dos fármacos , Privação do Sono/complicações , Animais , Cocaína/administração & dosagem , Transtornos Relacionados ao Uso de Cocaína/fisiopatologia , Inibidores da Captação de Dopamina/administração & dosagem , Masculino , Camundongos Endogâmicos C57BL , Privação do Sono/fisiopatologia
14.
Artigo em Inglês | MEDLINE | ID: mdl-31236515

RESUMO

Chronic cocaine use has been associated with sleep disturbances, both during active use periods and during withdrawal and abstinence. Acute cocaine also increases waking at the expense of slow wave sleep and Rapid Eye Movement in non-human subjects. However, the effects of acute cocaine on sleep/waking activity in mice, a rodent model commonly used in both sleep and addiction research due to its high genetic tractability, has yet to be investigated. Sleep/waking activity was measured via polysomnography following IP administration of three doses of cocaine (3.6, 9.6, 18 mg/kg) and vehicle control in male C57BL/6 mice. Cocaine dose-dependently increased sleep latency, increased waking time and increased fast EEG activity within waking. Increases in waking occurred primarily during the first hour following injection, followed by rebound SWS sleep. Sleep/waking activity normalized within a 24-hour period. As with humans and other rodents, cocaine dose dependently reduces sleep in a wildtype strain of mice commonly used in reward and addiction research.

15.
Curr Opin Neurobiol ; 44: 236-242, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28633050

RESUMO

Slow wave activity (SWA) during slow wave sleep (SWS) is the best indicator of the sleep homeostasis. The intensity of the SWA observed during SWS that follows prolonged waking is directly correlated with the duration of prior waking and its intensity decays during SWS suggesting a buildup and a resolution of sleep need. This sleep-homeostasis related SWA results from a buildup and decay of extracellular adenosine that acts at neuronal adenosine A1 receptors to facilitate SWA and is metabolized by adenosine kinase found in glia. This local neuronal-glial circuit for homeostatic SWA is primarily under the requisite control of two genes, the Adora1 and Adk, encoding the responsible adenosine receptor and adenosine's highest affinity metabolizing enzyme.


Assuntos
Adenosina/metabolismo , Homeostase/fisiologia , Sono/fisiologia , Homeostase/genética , Humanos , Neuroglia/fisiologia , Neurônios/fisiologia , Sono/genética
16.
Nature ; 537(7620): 357-362, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27602521

RESUMO

The retention of episodic-like memory is enhanced, in humans and animals, when something novel happens shortly before or after encoding. Using an everyday memory task in mice, we sought the neurons mediating this dopamine-dependent novelty effect, previously thought to originate exclusively from the tyrosine-hydroxylase-expressing (TH+) neurons in the ventral tegmental area. Here we report that neuronal firing in the locus coeruleus is especially sensitive to environmental novelty, locus coeruleus TH+ neurons project more profusely than ventral tegmental area TH+ neurons to the hippocampus, optogenetic activation of locus coeruleus TH+ neurons mimics the novelty effect, and this novelty-associated memory enhancement is unaffected by ventral tegmental area inactivation. Surprisingly, two effects of locus coeruleus TH+ photoactivation are sensitive to hippocampal D1/D5 receptor blockade and resistant to adrenoceptor blockade: memory enhancement and long-lasting potentiation of synaptic transmission in CA1 ex vivo. Thus, locus coeruleus TH+ neurons can mediate post-encoding memory enhancement in a manner consistent with possible co-release of dopamine in the hippocampus.


Assuntos
Dopamina/metabolismo , Locus Cerúleo/fisiologia , Consolidação da Memória/fisiologia , Animais , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/efeitos dos fármacos , Região CA1 Hipocampal/fisiologia , Técnicas In Vitro , Locus Cerúleo/citologia , Locus Cerúleo/efeitos da radiação , Masculino , Consolidação da Memória/efeitos dos fármacos , Consolidação da Memória/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Neurônios/efeitos da radiação , Optogenética , Receptores Adrenérgicos/metabolismo , Receptores de Dopamina D1/antagonistas & inibidores , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D5/antagonistas & inibidores , Receptores de Dopamina D5/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Área Tegmentar Ventral/citologia , Área Tegmentar Ventral/fisiologia
17.
J Neurosci ; 36(13): 3709-21, 2016 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-27030757

RESUMO

Sleep homeostasis reflects a centrally mediated drive for sleep, which increases during waking and resolves during subsequent sleep. Here we demonstrate that mice deficient for glial adenosine kinase (AdK), the primary metabolizing enzyme for adenosine (Ado), exhibit enhanced expression of this homeostatic drive by three independent measures: (1) increased rebound of slow-wave activity; (2) increased consolidation of slow-wave sleep; and (3) increased time constant of slow-wave activity decay during an average slow-wave sleep episode, proposed and validated here as a new index for homeostatic sleep drive. Conversely, mice deficient for the neuronal adenosine A1 receptor exhibit significantly decreased sleep drive as judged by these same indices. Neuronal knock-out of AdK did not influence homeostatic sleep need. Together, these findings implicate a glial-neuronal circuit mediated by intercellular Ado, controlling expression of homeostatic sleep drive. Because AdK is tightly regulated by glial metabolic state, our findings suggest a functional link between cellular metabolism and sleep homeostasis. SIGNIFICANCE STATEMENT: The work presented here provides evidence for an adenosine-mediated regulation of sleep in response to waking (i.e., homeostatic sleep need), requiring activation of neuronal adenosine A1 receptors and controlled by glial adenosine kinase. Adenosine kinase acts as a highly sensitive and important metabolic sensor of the glial ATP/ADP and AMP ratio directly controlling intracellular adenosine concentration. Glial equilibrative adenosine transporters reflect the intracellular concentration to the extracellular milieu to activate neuronal adenosine receptors. Thus, adenosine mediates a glial-neuronal circuit linking glial metabolic state to neural-expressed sleep homeostasis. This indicates a metabolically related function(s) for this glial-neuronal circuit in the buildup and resolution of our need to sleep and suggests potential therapeutic targets more directly related to sleep function.


Assuntos
Adenosina/metabolismo , Homeostase/fisiologia , Rede Nervosa/fisiologia , Neuroglia/fisiologia , Neurônios/fisiologia , Sono/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Adenosina Quinase/genética , Adenosina Quinase/imunologia , Adenosina Quinase/metabolismo , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Antagonistas de Estrogênios/farmacologia , Hipocampo/citologia , Hipocampo/fisiologia , Homeostase/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Receptor A1 de Adenosina/genética , Receptor A1 de Adenosina/metabolismo , Sono/genética , Tamoxifeno/farmacologia , Fatores de Tempo
18.
Hippocampus ; 25(11): 1374-9, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25786918

RESUMO

CA1 hippocampal N-methyl-d-aspartate-receptors (NMDARs) are necessary for contextually related learning and memory processes. Extinction, a form of learning, has been shown to require intact hippocampal NMDAR signalling. Renewal of fear expression can occur after fear extinction training, when the extinguished fear stimulus is presented in an environmental context different from the training context and thus, renewal is dependent on contextual memory. In this study, we show that a Grin1 knock-out (loss of the essential NR1 subunit for the NMDAR) restricted to the bilateral CA1 subfield of the dorsal hippocampus does not affect acquisition of learned fear, but does attenuate extinction of a cued fear response even when presented in the extinction-training context. We propose that failure to remember the (safe) extinction context is responsible for the abnormal fear response and suggest it is a dysfunctional renewal. The results highlight the difference in outcome of extinguished fear memory resulting from a partial rather than complete loss of function of the hippocampus and suggest a potential mechanism for abnormally increased fear expression in PTSD.


Assuntos
Comportamento Animal/fisiologia , Região CA1 Hipocampal/fisiopatologia , Extinção Psicológica/fisiologia , Medo/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Animais , Sinais (Psicologia) , Modelos Animais de Doenças , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Receptores de N-Metil-D-Aspartato/genética , Transtornos de Estresse Pós-Traumáticos/fisiopatologia
19.
Brain Res ; 1600: 70-83, 2015 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-25452020

RESUMO

Glutamate N-methyl-D-aspartate receptors (NMDARs) in the medial prefrontal cortex (mPFC) and hippocampus may play an integral role in complex cognitive and social deficits associated with a number of psychiatric illnesses including autism, mood disorders, and schizophrenia. We used localized infusions of adeno-associated virus Cre-recombinase in adult, targeted knock-in mice with loxP sites flanking exons 11-22 of the NR1 gene to investigate the effects of chronic NMDAR dysfunction in the mPFC and CA3 hippocampus on cognitive and social behavior. A 5-choice serial reaction time task (5-CSRTT) was used to monitor aspects of cognitive function that included attention and response inhibition. Social behavior was assessed using Crowley׳s sociability and preference for social novelty protocol. Chronic NMDAR dysfunction localized to the anterior cingulate/prelimbic mPFC or dorsal CA3 hippocampus differentially affected the response inhibition and social interaction. mPFC NR1-deletion increased perseverative responding in the 5-CSRTT and enhanced preference for social novelty, whereas CA3 NR1-deletion increased premature responding in the 5-CSRTT and decreased social approach behavior. These findings suggest that mPFC and CA3 NMDARs play selective roles in regulating compulsive and impulsive behavior, respectively. Furthermore, these findings are consistent with emerging evidence that these behaviors are mediated by distinct, albeit overlapping, neural circuits. Our data also suggest that NMDARs in these regions uniquely contribute to the expression of normal social behavior. In this case, mPFC and CA3 NMDARs appear to inhibit and facilitate aspects of social interaction, respectively. The latter dissociation raises the possibility that distinct circuits contribute to the expression of social intrusiveness and impoverished social interaction.


Assuntos
Cognição/fisiologia , Hipocampo/fisiologia , Comportamento Impulsivo/fisiologia , Córtex Pré-Frontal/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Comportamento Social , Animais , Deleção de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Tempo de Reação , Receptores de N-Metil-D-Aspartato/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...