Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 177
Filtrar
1.
Aging Cell ; 20(8): e13437, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34291567

RESUMO

Synapse degeneration correlates strongly with cognitive impairments in Alzheimer's disease (AD) patients. Soluble Amyloid-beta (Aß) oligomers are thought as the major trigger of synaptic malfunctions. Our earlier studies have demonstrated that Aß oligomers interfere with synaptic function through N-methyl-D-aspartate receptors (NMDARs). Our recent in vitro study found the neuroprotective role of astrocytic GluN2A in the promotion of synapse survival and identified nerve growth factor (NGF) derived from astrocytes, as a likely mediator of astrocytic GluN2A buffering against Aß synaptotoxicity. Our present in vivo study focused on exploring the precise mechanism of astrocytic GluN2A influencing Aß synaptotoxicity through regulating NGF. We generated an adeno-associated virus (AAV) expressing an astrocytic promoter (GfaABC1D) shRNA targeted to Grin2a (the gene encoding GluN2A) to perform astrocyte-specific Grin2a knockdown in the hippocampal dentate gyrus, after 3 weeks of virus vector expression, Aß were bilaterally injected into the intracerebral ventricle. Our results showed that astrocyte-specific knockdown of Grin2a and Aß application both significantly impaired spatial memory and cognition, which associated with the reduced synaptic proteins PSD95, synaptophysin and compensatory increased NGF. The reduced astrocytic GluN2A can counteract Aß-induced compensatory protective increase of NGF through regulating pNF-κB, Furin and VAMP3, which modulating the synthesis, mature and secretion of NGF respectively. Our present data reveal, for the first time, a novel mechanism of astrocytic GluN2A in exerting protective effects on synapses at the early stage of Aß exposure, which may contribute to establish new targets for AD prevention and early therapy.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Astrócitos/metabolismo , Transtornos Cognitivos/metabolismo , Disfunção Cognitiva/metabolismo , Fator de Crescimento Neural/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Astrócitos/patologia , Transtornos Cognitivos/patologia , Disfunção Cognitiva/patologia , Masculino , Ratos , Ratos Wistar , Receptores de N-Metil-D-Aspartato/deficiência , Receptores de N-Metil-D-Aspartato/genética
2.
Neuroimage ; 239: 118281, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34147627

RESUMO

Plasticity of synaptic strength and density is a vital mechanism enabling memory consolidation, learning, and neurodevelopment. It is strongly dependent on the intact function of N-Methyl-d-Aspartate Receptors (NMDAR). The importance of NMDAR is further evident as their dysfunction is involved in many diseases such as schizophrenia, Alzheimer's disease, neurodevelopmental disorders, and epilepsies. Synaptic plasticity is thought to be reflected by changes of sleep slow wave slopes across the night, namely higher slopes after wakefulness at the beginning of sleep than after a night of sleep. Hence, a functional NMDAR deficiency should theoretically lead to altered overnight changes of slow wave slopes. Here we investigated whether pediatric patients with anti-NMDAR encephalitis, being a very rare but unique human model of NMDAR deficiency due to autoantibodies against receptor subunits, indeed show alterations in this sleep EEG marker for synaptic plasticity. We retrospectively analyzed 12 whole-night EEGs of 9 patients (age 4.3-20.8 years, 7 females) and compared them to a control group of 45 healthy individuals with the same age distribution. Slow wave slopes were calculated for the first and last hour of Non-Rapid Eye Movement (NREM) sleep (factor 'hour') for patients and controls (factor 'group'). There was a significant interaction between 'hour' and 'group' (p = 0.013), with patients showing a smaller overnight decrease of slow wave slopes than controls. Moreover, we found smaller slopes during the first hour in patients (p = 0.022), whereas there was no group difference during the last hour of NREM sleep (p = 0.980). Importantly, the distribution of sleep stages was not different between the groups, and in our main analyses of patients without severe disturbance of sleep architecture, neither was the incidence of slow waves. These possible confounders could therefore not account for the differences in the slow wave slope values, which we also saw in the analysis of the whole sample of EEGs. These results suggest that quantitative EEG analysis of slow wave characteristics may reveal impaired synaptic plasticity in patients with anti-NMDAR encephalitis, a human model of functional NMDAR deficiency. Thus, in the future, the changes of sleep slow wave slopes may contribute to the development of electrophysiological biomarkers of functional NMDAR deficiency and synaptic plasticity in general.


Assuntos
Encefalite Antirreceptor de N-Metil-D-Aspartato/fisiopatologia , Ondas Encefálicas/fisiologia , Eletroencefalografia/métodos , Plasticidade Neuronal , Receptores de N-Metil-D-Aspartato/deficiência , Fases do Sono/fisiologia , Adolescente , Encefalite Antirreceptor de N-Metil-D-Aspartato/diagnóstico por imagem , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Receptores de N-Metil-D-Aspartato/imunologia , Estudos Retrospectivos , Adulto Jovem
3.
J Neurophysiol ; 126(1): 11-27, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34038186

RESUMO

There is substantial evidence that both N-methyl-D-aspartate receptor (NMDAR) hypofunction and dysfunction of GABAergic neurotransmission contribute to schizophrenia, though the relationship between these pathophysiological processes remains largely unknown. Although models using cell-type-specific genetic deletion of NMDARs have been informative, they display overly pronounced phenotypes extending beyond those of schizophrenia. Here, we used the serine racemase knockout (SRKO) mice, a model of reduced NMDAR activity rather than complete receptor elimination, to examine the link between NMDAR hypofunction and decreased GABAergic inhibition. The SRKO mice, in which there is a >90% reduction in the NMDAR coagonist d-serine, exhibit many of the neurochemical and behavioral abnormalities observed in schizophrenia. We found a significant reduction in inhibitory synapses onto CA1 pyramidal neurons in the SRKO mice. This reduction increases the excitation/inhibition balance resulting in enhanced synaptically driven neuronal excitability without changes in intrinsic excitability. Consistently, significant reductions in inhibitory synapse density in CA1 were observed by immunohistochemistry. We further show, using a single-neuron genetic deletion approach, that the loss of GABAergic synapses onto pyramidal neurons observed in the SRKO mice is driven in a cell-autonomous manner following the deletion of SR in individual CA1 pyramidal cells. These results support a model whereby NMDAR hypofunction in pyramidal cells disrupts GABAergic synapses leading to disrupted feedback inhibition and impaired neuronal synchrony.NEW & NOTEWORTHY Recently, disruption of excitation/inhibition (E/I) balance has become an area of considerable interest for psychiatric research. Here, we report a reduction in inhibition in the serine racemase knockout mouse model of schizophrenia that increases E/I balance and enhances synaptically driven neuronal excitability. This reduced inhibition was driven cell-autonomously in pyramidal cells lacking serine racemase, suggesting a novel mechanism for how chronic NMDA receptor hypofunction can disrupt information processing in schizophrenia.


Assuntos
Potenciais Pós-Sinápticos Excitadores/fisiologia , Neurônios GABAérgicos/metabolismo , Potenciais Pós-Sinápticos Inibidores/fisiologia , Racemases e Epimerases/deficiência , Receptores de N-Metil-D-Aspartato/deficiência , Sinapses/metabolismo , Animais , Feminino , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Técnicas de Cultura de Órgãos , Racemases e Epimerases/genética , Receptores de N-Metil-D-Aspartato/genética , Esquizofrenia/genética , Esquizofrenia/metabolismo , Sinapses/genética
4.
Addict Biol ; 26(6): e13042, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33864336

RESUMO

Astrocytes have become established as an important regulator of neuronal activity in the brain. Accumulating literature demonstrates that cocaine self-administration in rodent models induces structural changes within astrocytes that may influence their interaction with the surrounding neurons. Here, we provide evidence that cocaine impacts astrocytes at the functional level and alters neuronal sensitivity to astrocyte-derived glutamate. We report that a 14-day period of short access to cocaine (2 h/day) decreases spontaneous astrocytic Ca2+ transients and precipitates changes in astrocyte network activity in the nucleus accumbens shell. This is accompanied by increased prevalence of slow inward currents, a physiological marker of neuronal activation by astrocytic glutamate, in a subset of medium spiny neurons. Within, but not outside, of this subset, we observe an increase in duration and frequency of N-methyl-D-aspartate (NMDA) receptor-mediated synaptic events. Additionally, we find that the link between synaptic NMDA receptor plasticity and neuron sensitivity to astrocytic glutamate is maintained independent of drug exposure and is observed in both cocaine and saline control animals. Imaging analyses of neuronal Ca2+ activity show no effect of cocaine self-administration on individual cells or on neuronal network activity in brain slices. Therefore, our data indicate that cocaine self-administration promotes astrocyte-specific functional changes that can be linked to increased glutamate-mediated coupling with principal neurons in the nucleus accumbens. Such coupling may be spatially restricted as it does not result in a broad impact on network structure of local neuronal circuits.


Assuntos
Astrócitos/efeitos dos fármacos , Cocaína/farmacologia , Plasticidade Neuronal/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Animais , Astrócitos/metabolismo , Canais de Cálcio/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ácido Glutâmico/metabolismo , Masculino , Plasticidade Neuronal/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/deficiência , Autoadministração
5.
Schizophr Bull ; 47(5): 1300-1309, 2021 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-33822178

RESUMO

Altered Excitatory/Inhibitory (E/I) balance of cortical synaptic inputs has been proposed as a central pathophysiological factor for psychiatric neurodevelopmental disorders, including schizophrenia (SZ). However, direct measurement of E/I synaptic balance have not been assessed in vivo for any validated SZ animal model. Using a mouse model useful for the study of SZ we show that a selective ablation of NMDA receptors (NMDAr) in cortical and hippocampal interneurons during early postnatal development results in an E/I imbalance in vivo, with synaptic inputs to pyramidal neurons shifted towards excitation in the adult mutant medial prefrontal cortex (mPFC). Remarkably, this imbalance depends on the cortical state, only emerging when theta and gamma oscillations are predominant in the network. Additional brain slice recordings and subsequent 3D morphological reconstruction showed that E/I imbalance emerges after adolescence concomitantly with significant dendritic retraction and dendritic spine re-localization in pyramidal neurons. Therefore, early postnatal ablation of NMDAr in cortical and hippocampal interneurons developmentally impacts on E/I imbalance in vivo in an activity-dependent manner.


Assuntos
Ondas Encefálicas/fisiologia , Fenômenos Eletrofisiológicos/fisiologia , Hipocampo/fisiopatologia , Interneurônios/fisiologia , Rede Nervosa/fisiopatologia , Córtex Pré-Frontal/fisiopatologia , Células Piramidais/fisiologia , Receptores de N-Metil-D-Aspartato/deficiência , Esquizofrenia/fisiopatologia , Fatores Etários , Animais , Modelos Animais de Doenças , Hipocampo/metabolismo , Interneurônios/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Rede Nervosa/metabolismo , Parvalbuminas/metabolismo , Córtex Pré-Frontal/metabolismo , Células Piramidais/metabolismo , Esquizofrenia/metabolismo
6.
Cells ; 10(1)2021 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-33430405

RESUMO

Insulin-secreting pancreatic ß-cells express proteins characteristic of D-serine regulated synapses, but the acute effect of D-serine co-agonism on its presumptive ß-cell target, N-methyl D-aspartate receptors (NMDARs), is unclear. We used multiple models to evaluate glucose homeostasis and insulin secretion in mice with a systemic increase in D-serine (intraperitoneal injection or DAAO mutants without D-serine catabolism) or tissue-specific loss of Grin1-encoded GluN1, the D-serine binding NMDAR subunit. We also investigated the effects of D-serine ± NMDA on glucose-stimulated insulin secretion (GSIS) and ß-cell depolarizing membrane oscillations, using perforated patch electrophysiology, in ß-cell-containing primary isolated mouse islets. In vivo models of elevated D-serine correlated to improved blood glucose and insulin levels. In vitro, D-serine potentiated GSIS and ß-cell membrane excitation, dependent on NMDAR activating conditions including GluN1 expression (co-agonist target), simultaneous NMDA (agonist), and elevated glucose (depolarization). Pancreatic GluN1-loss females were glucose intolerant and GSIS was depressed in islets from younger, but not older, ßGrin1 KO mice. Thus, D-serine is capable of acute antidiabetic effects in mice and potentiates insulin secretion through excitatory ß-cell NMDAR co-agonism but strain-dependent shifts in potency and age/sex-specific Grin1-loss phenotypes suggest that context is critical to the interpretation of data on the role of D-serine and NMDARs in ß-cell function.


Assuntos
Glucose/farmacologia , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Receptores de N-Metil-D-Aspartato/agonistas , Serina/metabolismo , Animais , Glicemia/metabolismo , Modelos Animais de Doenças , Feminino , Intolerância à Glucose/metabolismo , Homeostase , Células Secretoras de Insulina/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , N-Metilaspartato/metabolismo , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/metabolismo , Fenótipo , Receptores de N-Metil-D-Aspartato/deficiência , Receptores de N-Metil-D-Aspartato/metabolismo , Caracteres Sexuais
7.
J Neurosci ; 41(6): 1207-1217, 2021 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-33372060

RESUMO

Correlated spontaneous activity plays critical role in the organization of neocortical circuits during development. However, cortical mechanisms regulating activity correlation are still elusive. In this study, using two-photon calcium imaging of the barrel cortex layer 4 (L4) in living neonatal mice, we found that NMDA receptors (NMDARs) in L4 neurons are important for enhancement of spontaneous activity correlation. Disruption of GluN1 (Grin1), an obligatory NMDAR subunit, in a sparse population of L4 neurons reduced activity correlation between GluN1 knock-out (GluN1KO) neuron pairs within a barrel. This reduction in activity correlation was even detected in L4 neuron pairs in neighboring barrels and most evident when either or both of neurons are located on the barrel edge. Our results provide evidence for the involvement of L4 neuron NMDARs in spatial organization of the spontaneous firing activity of L4 neurons in the neonatal barrel cortex.SIGNIFICANCE STATEMENT Precise wiring of the thalamocortical circuits is necessary for proper sensory information processing, and thalamus-derived correlated spontaneous activity is important for thalamocortical circuit formation. The molecular mechanisms involved in the correlated activity transfer from the thalamus to the neocortex are largely unknown. In vivo two-photon calcium imaging of the neonatal barrel cortex revealed that correlated spontaneous activity between layer four neurons is reduced by mosaic knock-out (KO) of the NMDA receptor (NMDAR) obligatory subunit GluN1. Our results suggest that the function of NMDARs in layer four neurons is necessary for the communication between presynaptic and postsynaptic partners during thalamocortical circuit formation.


Assuntos
Potenciais Pós-Sinápticos Excitadores/fisiologia , Proteínas do Tecido Nervoso/deficiência , Receptores de N-Metil-D-Aspartato/deficiência , Córtex Somatossensorial/citologia , Córtex Somatossensorial/metabolismo , Animais , Animais Recém-Nascidos , Feminino , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Imagem Molecular/métodos , Proteínas do Tecido Nervoso/genética , Receptores de N-Metil-D-Aspartato/genética
8.
J Neurosci ; 40(47): 9121-9136, 2020 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-33051351

RESUMO

Abnormalities in interactions between sensory neurons and Schwann cells (SCs) may result in heightened pain processing and chronic pain states. We previously reported that SCs express the NMDA receptor (NMDA-R), which activates cell signaling in response to glutamate and specific protein ligands, such as tissue-type plasminogen activator. Herein, we genetically targeted grin1 encoding the essential GluN1 NMDA-R subunit, conditionally in SCs, to create a novel mouse model in which SCs are NMDA-R-deficient (GluN1- mice). These mice demonstrated increased sensitivity to light touch, pinprick, and thermal hyperalgesia in the absence of injury, without associated changes in motor function. Ultrastructural analysis of adult sciatic nerve in GluN1- mice revealed increases in the density of Aδ fibers and Remak bundles and a decrease in the density of Aß fibers, without altered g-ratios. Abnormalities in adult Remak bundle ultrastructure were also present including aberrant C-fiber ensheathment, distances between axons, and increased poly-axonal pockets. Developmental and post radial sorting defects contributed to altered nerve fiber densities in adult. Uninjured sciatic nerves in GluN1- mice did not demonstrate an increase in neuroinflammatory infiltrates. Transcriptome profiling of dorsal root ganglia (DRGs) revealed 138 differentially regulated genes in GluN1- mice. One third of the regulated genes are known to be involved in pain processing, including sprr1a, npy, fgf3, atf3, and cckbr, which were significantly increased. The intraepidermal nerve fiber density (IENFD) was significantly decreased in the skin of GluN1- mice. Collectively, these findings demonstrate that SC NMDA-R is essential for normal PNS development and for preventing development of pain states.SIGNIFICANCE STATEMENT Chronic unremitting pain is a prevalent medical condition; however, the molecular mechanisms that underlie heightened pain processing remain incompletely understood. Emerging data suggest that abnormalities in Schwann cells (SCs) may cause neuropathic pain. We established a novel mouse model for small fiber neuropathy (SFN) in which grin1, the gene that encodes the NMDA receptor (NMDA-R) GluN1 subunit, is deleted in SCs. These mice demonstrate hypersensitivity in pain processing in the absence of nerve injury. Changes in the density of intraepidermal small fibers, the ultrastructure of Remak bundles, and the transcriptome of dorsal root ganglia (DRGs) provide possible explanations for the increase in pain processing. Our results support the hypothesis that abnormalities in communication between sensory nerve fibers and SCs may result in pain states.


Assuntos
Hiperalgesia/genética , Proteínas do Tecido Nervoso/genética , Dor/genética , Dor/fisiopatologia , Receptores de N-Metil-D-Aspartato/genética , Células de Schwann/ultraestrutura , Animais , Axônios/fisiologia , Axônios/ultraestrutura , Gânglios Espinais/citologia , Gânglios Espinais/fisiologia , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fibras Nervosas/fisiologia , Proteínas do Tecido Nervoso/deficiência , Estimulação Física , Cultura Primária de Células , Receptores de N-Metil-D-Aspartato/deficiência , Nervo Isquiático/ultraestrutura , Transdução de Sinais
9.
Mol Psychiatry ; 25(11): 2832-2843, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-30038231

RESUMO

Recent findings from in vivo-imaging and human post-mortem tissue studies in schizophrenic psychosis (SzP), have demonstrated functional and molecular changes in hippocampal subfields that can be associated with hippocampal hyperexcitability. In this study, we used a subfield-specific GluN1 knockout mouse with a disease-like molecular perturbation expressed only in hippocampal dentate gyrus (DG) and assessed its association with hippocampal physiology and psychosis-like behaviors. First, we used whole-cell patch-clamp recordings to measure the physiological changes in hippocampal subfields and cFos immunohistochemistry to examine cellular excitability. DG-GluN1 KO mice show CA3 cellular hyperactivity, detected using two approaches: (1) increased excitatory glutamate transmission at mossy fibers (MF)-CA3 synapses, and (2) an increased number of cFos-activated pyramidal neurons in CA3, an outcome that appears to project downstream to CA1 and basolateral amygdala (BLA). Furthermore, we examined psychosis-like behaviors and pathological memory processing; these show an increase in fear conditioning (FC), a reduction in prepulse inhibition (PPI) in the KO animal, along with a deterioration in memory accuracy with Morris Water Maze (MWM) and reduced social memory (SM). Moreover, with DREADD vectors, we demonstrate a remarkably similar behavioral profile when we induce CA3 hyperactivity. These hippocampal subfield changes could provide the basis for the observed increase in human hippocampal activity in SzP, based on the shared DG-specific GluN1 reduction. With further characterization, these animal model systems may serve as targets to test psychosis mechanisms related to hippocampus and assess potential hippocampus-directed treatments.


Assuntos
Região CA3 Hipocampal/fisiopatologia , Giro Denteado/metabolismo , Proteínas do Tecido Nervoso/deficiência , Transtornos Psicóticos/fisiopatologia , Receptores de N-Metil-D-Aspartato/deficiência , Animais , Região CA3 Hipocampal/citologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células Piramidais
10.
Cereb Cortex ; 30(4): 2555-2572, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-31832634

RESUMO

Primary visual cortex (V1) is the locus of numerous forms of experience-dependent plasticity. Restricting visual stimulation to one eye at a time has revealed that many such forms of plasticity are eye-specific, indicating that synaptic modification occurs prior to binocular integration of thalamocortical inputs. A common feature of these forms of plasticity is the requirement for NMDA receptor (NMDAR) activation in V1. We therefore hypothesized that NMDARs in cortical layer 4 (L4), which receives the densest thalamocortical input, would be necessary for all forms of NMDAR-dependent and input-specific V1 plasticity. We tested this hypothesis in awake mice using a genetic approach to selectively delete NMDARs from L4 principal cells. We found, unexpectedly, that both stimulus-selective response potentiation and potentiation of open-eye responses following monocular deprivation (MD) persist in the absence of L4 NMDARs. In contrast, MD-driven depression of deprived-eye responses was impaired in mice lacking L4 NMDARs, as was L4 long-term depression in V1 slices. Our findings reveal a crucial requirement for L4 NMDARs in visual cortical synaptic depression, and a surprisingly negligible role for them in cortical response potentiation. These results demonstrate that NMDARs within distinct cellular subpopulations support different forms of experience-dependent plasticity.


Assuntos
Potenciais Evocados Visuais/fisiologia , Plasticidade Neuronal/fisiologia , Estimulação Luminosa/métodos , Receptores de N-Metil-D-Aspartato/deficiência , Privação Sensorial/fisiologia , Córtex Visual/fisiologia , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptores de N-Metil-D-Aspartato/genética
11.
Cell Rep ; 29(13): 4285-4294.e5, 2019 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-31875540

RESUMO

NMDA receptors (NMDARs) are critical for physiological synaptic plasticity, learning, and memory and for pathological plasticity and neuronal death. The GluN1 subunit is encoded by a single gene, GRIN1, with 8 splice variants, but whether the diversity generated by this splicing has physiological consequences remains enigmatic. Here, we generate mice lacking from the GluN1 exon 5-encoded N1 cassette (GluN1a mice) or compulsorily expressing this exon (GluN1b mice). Despite no differences in basal synaptic transmission, long-term potentiation in the hippocampus is significantly enhanced in GluN1a mice compared with that in GluN1b mice. Furthermore, GluN1a mice learn more quickly and have significantly better spatial memory performance than do GluN1b mice. In addition, in human iPSC-derived neurons in autism spectrum disorder NMDARs show characteristics of N1-lacking GluN1. Our findings indicate that alternative splicing of GluN1 is a mechanism for controlling physiological long-lasting synaptic potentiation, learning, and memory.


Assuntos
Processamento Alternativo , Transtorno do Espectro Autista/genética , Potenciação de Longa Duração/genética , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Animais , Transtorno do Espectro Autista/metabolismo , Transtorno do Espectro Autista/patologia , Morte Celular/genética , Diferenciação Celular , Potenciais Pós-Sinápticos Excitadores/fisiologia , Éxons , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência , Neurônios/patologia , Cultura Primária de Células , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética , Receptores de N-Metil-D-Aspartato/deficiência , Memória Espacial/fisiologia , Sinapses/metabolismo , Transmissão Sináptica
12.
Neuroscience ; 415: 18-30, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31325561

RESUMO

Previous studies have focused on the effects of N-methyl-D-aspartate receptor (NMDAR) blockade on neonates, but little is known about the effect of the embryonic NMDAR blockade on offspring, especially the long-lasting effect, on behavior in adulthood. Here, pregnant rats at E14 were treated with ketamine for 5 successive days and undergone multiple behavior tests, electrophysiology experiment, and Western blotting analysis to detect the alterations in their offspring. We found that embryonic ketamine treatment induced anxiety-like behavior in adulthood (8-week old) offspring. At the same period, we observed an attenuation of NMDA-evoked current as well as decreased NR2A and NR2B membrane expression in the prefrontal cortex (PFC), but not in the hippocampus or amygdala. Selective inhibition experiments with NR2A or NR2B specific antagonists suggested that embryonic ketamine treatment induced NMDAR current attenuation was likely mediated by changes in NR2A subunit. Moreover, at the 4-week time point, NMDA-evoked current was unchanged in PFC, but enhanced in hippocampal CA1 area, which may be caused by the over expression of NR2B in the hippocampus at 4-week time. Furthermore, NR2B knockdown, by using NR2B-shRNA lentivirus, in the hippocampal CA1 area at 3-4-week of age significantly rescued the decrease in NR2A expression in the PFC and anxiety-like behavior observed at 8-week adult offspring rats. In conclusion, our results suggested that embryonic ketamine treatment induced anxiety-like behavior and the downregulation of NMDAR function in PFC in the adulthood period of offspring, which might result from the enhanced function of NMDARs in the hippocampus at the 4-week juvenile time point.


Assuntos
Ansiedade/induzido quimicamente , Antagonistas de Aminoácidos Excitatórios/farmacologia , Ketamina/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Regulação para Baixo , Feminino , Elevação dos Membros Posteriores , Hipocampo/metabolismo , Hipocampo/fisiologia , Masculino , Córtex Pré-Frontal/fisiologia , Gravidez , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/deficiência
13.
Mol Neurobiol ; 56(11): 7473-7489, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31044365

RESUMO

Kainate type ionotropic glutamate receptors (KARs) are expressed in hippocampal interneurons and regulate interneuron excitability and GABAergic transmission. Neuropilin tolloid-like proteins (NETO1 and NETO2) act as KAR auxiliary subunits; however, their significance for various functions of KARs in GABAergic interneurons is not fully understood. Here we show that NETO1, but not NETO2, is necessary for dendritic delivery of KAR subunits and, consequently, for formation of KAR-containing synapses in cultured GABAergic neurons. Accordingly, electrophysiological analysis of neonatal CA3 stratum radiatum interneurons revealed impaired postsynaptic and metabotropic KAR signaling in Neto1 knockouts, while a subpopulation of ionotropic KARs in the somatodendritic compartment remained functional. Loss of NETO1/KAR signaling had no significant effect on development of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and N-methyl-D-aspartate (NMDA)-receptor-mediated glutamatergic transmission in CA3 interneurons, contrasting the synaptogenic role proposed for KARs in principal cells. Furthermore, loss of NETO1 had no effect on excitability and characteristic spontaneous network bursts in the immature CA3 circuitry. However, we find that NETO1 is critical for kainate-dependent modulation of network bursts and GABAergic transmission in the hippocampus already during the first week of life. Our results provide the first description of NETO1-dependent subcellular targeting of KAR subunits in GABAergic neurons and indicate that endogenous NETO1 is required for formation of KAR-containing synapses in interneurons. Since aberrant KAR-mediated excitability is implicated in certain forms of epilepsy, NETO1 represents a potential therapeutic target for treatment of both adult and early life seizures.


Assuntos
Região CA3 Hipocampal/metabolismo , Interneurônios/metabolismo , Rede Nervosa/metabolismo , Receptores de Ácido Caínico/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/metabolismo , Potenciais de Ação , Animais , Axônios/metabolismo , Dendritos/metabolismo , Neurônios GABAérgicos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Subunidades Proteicas/metabolismo , Receptores de N-Metil-D-Aspartato/deficiência
14.
Neurosci Lett ; 699: 109-114, 2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-30726715

RESUMO

The dopamine (DA) system is critical for various forms of learning about salient environmental stimuli. Prior work has shown that deletion of the obligatory NR1 subunit of the N-methyl-D-aspartate (NMDA) receptor on neurons expressing the DA transporter (DAT) in mice results in reduced phasic release from DA-containing neurons. To further investigate the contribution of phasic DA release to reward-related learning and cognitive flexibility, the current study evaluated DAT-NR1 null mutant mice in a touchscreen-based pairwise visual discrimination and reversal learning paradigm. Results showed that these mutants were slower to attain a high level of choice accuracy on the discrimination task, but showed improved late reversal performance on sessions where correct choice was above chance. A number of possible interpretations are offered for this pattern of effects, including the opposing possibilities that discrimination memory was either stronger by the completion of training (overtraining effect) or weaker (learning deficit), both of which could potentially produce faster reversal. These data add to the extensive literature ascribing a critical role for DAergic neurotransmission in cognitive functions and the regulation of reward-related behaviors of relevance to addictions.


Assuntos
Discriminação Psicológica/fisiologia , Neurônios Dopaminérgicos/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Reversão de Aprendizagem/fisiologia , Animais , Feminino , Masculino , Camundongos , Camundongos Mutantes Neurológicos , Estimulação Luminosa , Receptores de N-Metil-D-Aspartato/deficiência , Percepção Visual/fisiologia
15.
Neuroscience ; 404: 338-352, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30742964

RESUMO

A common feature across neuropsychiatric disorders is inability to discontinue an action or thought once it has become detrimental. Reversal learning, a hallmark of executive control, requires plasticity within cortical, striatal and limbic circuits and is highly sensitive to disruption of N-methyl-D-aspartate receptor (NMDAR) function. In particular, selective deletion or antagonism of GluN2B containing NMDARs in cortical regions including the orbitofrontal cortex (OFC), promotes maladaptive perseveration. It remains unknown whether GluN2B functions to maintain local cortical activity necessary for reversal learning, or if it exerts a broader influence on the integration of neural activity across cortical and subcortical systems. To address this question, we utilized in vivo electrophysiology to record neuronal activity and local field potentials (LFP) in the orbitofrontal cortex and dorsal striatum (dS) of mice with deletion of GluN2B in neocortical and hippocampal principal cells while they performed touchscreen reversal learning. Reversal impairment produced by corticohippocampal GluN2B deletion was paralleled by an aberrant increase in functional connectivity between the OFC and dS. These alterations in coordination were associated with alterations in local OFC and dS firing activity. These data demonstrate highly dynamic patterns of cortical and striatal activity concomitant with reversal learning, and reveal GluN2B as a molecular mechanism underpinning the timing of these processes.


Assuntos
Disfunção Cognitiva/metabolismo , Corpo Estriado/metabolismo , Córtex Pré-Frontal/metabolismo , Receptores de N-Metil-D-Aspartato/deficiência , Reversão de Aprendizagem/fisiologia , Animais , Disfunção Cognitiva/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores de N-Metil-D-Aspartato/genética
16.
eNeuro ; 5(4)2018.
Artigo em Inglês | MEDLINE | ID: mdl-30302389

RESUMO

Selecting the most advantageous actions in a changing environment is a central feature of adaptive behavior. The midbrain dopamine (DA) neurons along with the major targets of their projections, including dopaminoceptive neurons in the frontal cortex and basal ganglia, play a key role in this process. Here, we investigate the consequences of a selective genetic disruption of NMDA receptor and metabotropic glutamate receptor 5 (mGluR5) in the DA system on adaptive choice behavior in mice. We tested the effects of the mutation on performance in the probabilistic reinforcement learning and probability-discounting tasks. In case of the probabilistic choice, both the loss of NMDA receptors in dopaminergic neurons or the loss mGluR5 receptors in D1 receptor-expressing dopaminoceptive neurons reduced the probability of selecting the more rewarded alternative and lowered the likelihood of returning to the previously rewarded alternative (win-stay). When observed behavior was fitted to reinforcement learning models, we found that these two mutations were associated with a reduced effect of the expected outcome on choice (i.e., more random choices). None of the mutations affected probability discounting, which indicates that all animals had a normal ability to assess probability. However, in both behavioral tasks animals with targeted loss of NMDA receptors in dopaminergic neurons or mGluR5 receptors in D1 neurons were significantly slower to perform choices. In conclusion, these results show that glutamate receptor-dependent signaling in the DA system is essential for the speed and accuracy of choices, but at the same time probably is not critical for correct estimation of probable outcomes.


Assuntos
Comportamento Animal/fisiologia , Tomada de Decisões/fisiologia , Dopamina/metabolismo , Neurônios/metabolismo , Receptor de Glutamato Metabotrópico 5/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Reforço Psicológico , Transdução de Sinais/fisiologia , Animais , Neurônios Dopaminérgicos/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Receptor de Glutamato Metabotrópico 5/deficiência , Receptores de N-Metil-D-Aspartato/deficiência , Recompensa
17.
Neuropsychopharmacology ; 43(12): 2383-2389, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29982266

RESUMO

Transient upregulation of GluN2B-containing NMDA receptors (R) in the nucleus accumbens (NAc) is proposed as an intermediate to long-term AMPAR plasticity associated with persistent cocaine-related behaviors. However, cell type- and input-specific contributions of GluN2B underlying lasting actions of cocaine remain to be elucidated. We utilized GluN2B cell type-specific knockouts and optogenetics to deconstruct the role of GluN2B in cocaine-induced NAc synaptic and behavioral plasticity. While reward learning was unaffected, loss of GluN2B in D1 dopamine receptor-expressing cells (D1) led to prolonged retention of reward memory. In control mice, prefrontal cortex (PFC)-D1(+) NAc AMPAR function was unaffected by cocaine exposure, while midline thalamus (mThal)-D1(+) NAc AMPAR function was potentiated but diminished after withdrawal. In D1-GluN2B-/- mice, the potentiation of mThal-D1(+) NAc AMPAR function persisted following withdrawal, corresponding with continued expression of cocaine reward behavior. These data suggest NAc GluN2B-containing NMDARs serve a feedback role and may weaken reward-related memories.


Assuntos
Cocaína/administração & dosagem , Núcleo Accumbens/metabolismo , Receptores de Dopamina D1/biossíntese , Receptores de N-Metil-D-Aspartato/deficiência , Recompensa , Tálamo/metabolismo , Animais , Deleção de Genes , Locomoção/efeitos dos fármacos , Locomoção/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Núcleo Accumbens/efeitos dos fármacos , Receptores de Dopamina D1/genética , Receptores de N-Metil-D-Aspartato/genética , Tálamo/efeitos dos fármacos
18.
Cell ; 174(1): 32-43.e15, 2018 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-29958111

RESUMO

The organization of action into sequences underlies complex behaviors that are essential for organismal survival and reproduction. Despite extensive studies of innate sequences in relation to central pattern generators, how learned action sequences are controlled and whether they are organized as a chain or a hierarchy remain largely unknown. By training mice to perform heterogeneous action sequences, we demonstrate that striatal direct and indirect pathways preferentially encode different behavioral levels of sequence structure. State-dependent closed-loop optogenetic stimulation of the striatal direct pathway can selectively insert a single action element into the sequence without disrupting the overall sequence length. Optogenetic manipulation of the striatal indirect pathway completely removes the ongoing subsequence while leaving the following subsequence to be executed with the appropriate timing and length. These results suggest that learned action sequences are not organized in a serial but rather a hierarchical structure that is distinctly controlled by basal ganglia pathways.


Assuntos
Aprendizagem , Neurônios/metabolismo , Optogenética , Animais , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/efeitos da radiação , Toxina Diftérica/farmacologia , Eletrodos Implantados , Potenciais Evocados Visuais , Feminino , Lasers , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Muscimol/farmacologia , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Proteínas RGS/genética , Receptores de N-Metil-D-Aspartato/deficiência , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo
19.
Behav Brain Res ; 353: 124-128, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-29953905

RESUMO

The N-methyl-D-aspartate receptor (NMDAR) is mechanistically involved in the behavioral and neurophysiological effects of alcohol, but the specific role of the GluN2A subunit remains unclear. Here, we exposed mice with constitutive GluN2A gene knockout (KO) to chronic intermittent ethanol vapor (CIE) and tested for EtOH consumption/preference using a two-bottle choice paradigm, as well as NMDAR-mediated transmission at basolateral amygdala synapses via ex vivo slice electrophysiology. Results showed that GluN2A KO mice attained comparable blood EtOH levels in response to CIE exposure, but did not exhibit the significant increase in EtOH drinking that was observed in CIE-exposed wildtypes. GluN2A KO mice also showed no alterations in BLA NMDAR-mediated synaptic transmission after CIE, relative to air-exposed, whereas C57BL/6 J mice showed an attenuated synaptic response to GluN2B antagonism. Taken together, these data add to mounting evidence supporting GluN2A-containing NMDARs as a mechanism underlying relative risk for developing EtOH dependence after repeated EtOH exposure.


Assuntos
Alcoolismo/metabolismo , Receptores de N-Metil-D-Aspartato/deficiência , Consumo de Bebidas Alcoólicas/metabolismo , Animais , Complexo Nuclear Basolateral da Amígdala/efeitos dos fármacos , Complexo Nuclear Basolateral da Amígdala/metabolismo , Depressores do Sistema Nervoso Central/administração & dosagem , Comportamento de Escolha/fisiologia , Etanol/administração & dosagem , Feminino , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fenóis/farmacologia , Piperidinas/farmacologia , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/genética , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Técnicas de Cultura de Tecidos
20.
Exp Neurol ; 305: 1-12, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29554474

RESUMO

The N-methyl-d-aspartate receptor (NMDAR) has been implicated in the pathophysiology of neurological diseases, such as schizophrenia, autism spectrum disorders (ASD), and Alzheimer's disease (AD), whose unique clinical hallmark is a constellation of impaired social and/or cognitive behaviors. GluN3A (NR3A) is a unique inhibitory subunit in the NMDAR complex. The role of GluN3A in social behavioral activities is obscure. In this study, we sought to evaluate altered social activities in adult GluN3A knockout (KO) mice. GluN3A KO mice spent less time in reciprocal social interaction in the social interaction test compared to wild-type (WT) mice. A social approach test using a three-chamber system confirmed that mice lacking GluN3A had lower sociability and did not exhibit a preference for social novelty. GluN3A KO mice displayed abnormal food preference in the social transmission of food preference task and low social interaction activity in the five-trial social memory test, but without social memory deficits. Using a home cage monitoring system, we observed reduced social grooming behavior in GluN3A KO mice. Signaling genes that might mediate the altered social behaviors were examined in the prefrontal cortex, hippocampus, and thalamus. Among nine genes examined, the expression of the oxytocin receptor was significantly lower in the prefrontal cortex of GluN3A KO mice than that in WT mice. Oxytocin treatment rescued social activity deficits in GluN3A KO mice. These findings support a novel idea that a chronic state of moderate increases in NMDAR activities may lead to downregulation of the oxytocin signaling and impaired behavioral activities that are seen in psychiatric/neurodegenerative disorders.


Assuntos
Asseio Animal/fisiologia , Receptores de N-Metil-D-Aspartato/deficiência , Receptores de Ocitocina/biossíntese , Transdução de Sinais/fisiologia , Comportamento Social , Fatores Etários , Animais , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ocitocina/biossíntese , Córtex Pré-Frontal/metabolismo , Subunidades Proteicas/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...