Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
1.
J Biol Chem ; 300(5): 107253, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38569938

RESUMO

Homocysteine, a sulfur-containing amino acid derived from methionine metabolism, is a known agonist of N-methyl-D-aspartate receptor (NMDAR) and is involved in neurotoxicity. Our previous findings showed that neuronal exposure to elevated homocysteine levels leads to sustained low-level increase in intracellular Ca2+, which is dependent on GluN2A subunit-containing NMDAR (GluN2A-NMDAR) stimulation. These studies further showed a role of ERK MAPK in homocysteine-GluN2A-NMDAR-mediated neuronal death. However, the intracellular mechanisms associated with such sustained GluN2A-NMDAR stimulation and subsequent Ca2+ influx have remained unexplored. Using live-cell imaging with Fluo3-AM and biochemical approaches, we show that homocysteine-GluN2A NMDAR-induced initial Ca2+ influx triggers sequential phosphorylation and subsequent activation of the proline rich tyrosine kinase 2 (Pyk2) and Src family kinases, which in turn phosphorylates GluN2A-Tyr1325 residue of GluN2A-NMDARs to maintain channel activity. The continuity of this cycle of events leads to sustained Ca2+ influx through GluN2A-NMDAR. Our findings also show that lack of activation of the regulatory tyrosine phosphatase STEP, which can limit Pyk2 and Src family kinase activity further contributes to the maintenance of this cycle. Additional studies using live-cell imaging of neurons expressing a redox-sensitive GFP targeted to the mitochondrial matrix show that treatment with homocysteine leads to a progressive increase in mitochondrial reactive oxygen species generation, which is dependent on GluN2A-NMDAR-mediated sustained ERK MAPK activation. This later finding demonstrates a novel role of GluN2A-NMDAR in homocysteine-induced mitochondrial ROS generation and highlights the role of ERK MAPK as the intermediary signaling pathway between GluN2A-NMDAR stimulation and mitochondrial reactive oxygen species generation.


Assuntos
Homocisteína , Mitocôndrias , Espécies Reativas de Oxigênio , Receptores de N-Metil-D-Aspartato , Receptores de N-Metil-D-Aspartato/metabolismo , Homocisteína/metabolismo , Homocisteína/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Animais , Mitocôndrias/metabolismo , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Cálcio/metabolismo , Fosforilação/efeitos dos fármacos , Quinase 2 de Adesão Focal/metabolismo , Quinases da Família src/metabolismo , Ratos , Camundongos , Humanos
2.
Int J Neuropsychopharmacol ; 27(9)2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-39185814

RESUMO

BACKGROUND: Depression is a heterogeneous disorder with high morbidity and disability rates that poses serious problems regarding mental health care. It is now well established that N-methyl D-aspartate receptor (NMDAR) modulators are being increasingly explored as potential therapeutic options for treating depression, although relatively little is known about their mechanisms of action. NMDARs are glutamate-gated ion channels that are ubiquitously expressed in the central nervous system (CNS), and they have been shown to play key roles in excitatory synaptic transmission. GluN2A, the predominant Glu2N subunit of functional NMDARs in neurons, is involved in various physiological processes in the CNS and is associated with diseases such as anxiety, depression, and schizophrenia. However, the role of GluN2A in the pathophysiology of depression has not yet been elucidated. METHODS: We reviewed several past studies to better understand the function of GluN2A in depression. Additionally, we also summarized the pathogenesis of depression based on the regulation of GluN2A expression, particularly its interaction with neuroinflammation and neurogenesis, which has received considerable critical attention and is highly implicated in the onset of depression. RESULTS: These evidence suggests that GluN2A overexpression impairs structural and functional synaptic plasticity, which contributes to the development of depression. Consequently, this knowledge is vital for the development of selective antagonists targeting GluN2A subunits using pharmacological and molecular methods. CONCLUSIONS: Specific inhibition of the GluN2A NMDAR subunit is resistant to chronic stress-induced depressive-like behaviors, making them promising targets for the development of novel antidepressants.


Assuntos
Antidepressivos , Receptores de N-Metil-D-Aspartato , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Humanos , Antidepressivos/farmacologia , Animais , Depressão/tratamento farmacológico , Depressão/metabolismo , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Transtorno Depressivo/tratamento farmacológico , Transtorno Depressivo/metabolismo , Desenvolvimento de Medicamentos , Neurogênese/efeitos dos fármacos
3.
Int J Mol Sci ; 25(18)2024 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-39337704

RESUMO

N-methyl-D-aspartate receptors (NMDARs) are glutamate receptors with key roles in synaptic communication and plasticity. The activation of synaptic NMDARs initiates plasticity and stimulates cell survival. In contrast, the activation of extrasynaptic NMDARs can promote cell death underlying a potential mechanism of neurodegeneration occurring in Alzheimer's disease (AD). The distribution of synaptic versus extrasynaptic NMDARs has emerged as an important parameter contributing to neuronal dysfunction in neurodegenerative diseases including AD. Here, we review the concept of extrasynaptic NMDARs, as this population is present in numerous neuronal cell membranes but also in the membranes of various non-neuronal cells. Previous evidence regarding the membranal distribution of synaptic versus extrasynaptic NMDRs in relation to AD mice models and in the brains of AD patients will also be reviewed.


Assuntos
Doença de Alzheimer , Receptores de N-Metil-D-Aspartato , Sinapses , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Humanos , Animais , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/metabolismo , Sinapses/patologia , Membrana Celular/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Plasticidade Neuronal , Membranas Sinápticas/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia
4.
Cell Mol Neurobiol ; 43(1): 139-153, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34978648

RESUMO

NMDA receptors play an important physiological role in regulating synaptic plasticity, learning and memory. GluN2A subunits are the most abundant functional subunits of NMDA receptors expressed in mature brain, and their dysfunction is related to various neurological diseases. According to subunit composition, GluN2A-containing NMDA receptors can be divided into two types: diheteromeric and triheteromeric receptors. In this review, the expression, functional and pharmacological properties of different kinds of GluN2A-containing NMDA receptors as well as selective GluN2A regulators were described to further understand this type of NMDA receptors.


Assuntos
Plasticidade Neuronal , Receptores de N-Metil-D-Aspartato , Receptores de N-Metil-D-Aspartato/metabolismo , Antagonistas de Aminoácidos Excitatórios/farmacologia
5.
Bioorg Med Chem ; 83: 117236, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36934527

RESUMO

NMDA receptor subunits have differential roles in mediating excitotoxic neuronal death both in vitro and in vivo . Activation of NR2A-containing NMDA receptors promotes neuronal survival and exerts a neuroprotective action, whereas over activating GluN2B-containing receptor results in excitotoxicity, increasing neuronal apoptosis. Our previous study has identified Npam 43 as a NMDAR positive allosteric modulators. However, the cis-trans isomerization impedes the development of Npam 43 as potential neuroprotective agents. To discover more potent and selective GluN2A NMDAR positive allosteric modulators, 38 derivatives were synthesized and evaluated their neuroprotective effect on glutamate-exposed PC-12 cells. The allosteric activities of compounds were evaluated using calcium imaging approaches. Among them, compound 5c exhibit GluN1/2A selectivity over GluN1/2B and show neuroprotective activity in vitro and in vivo. This study reported a series of GluN1/2A positive allosteric modulators as neuroprotective agents, and provided a potential opportunity to discover new drugs for stroke treatment.


Assuntos
Fármacos Neuroprotetores , Receptores de N-Metil-D-Aspartato , Apoptose , Morte Celular , Fármacos Neuroprotetores/farmacologia , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais , Acidente Vascular Cerebral/tratamento farmacológico , Regulação Alostérica/efeitos dos fármacos
6.
Bioorg Med Chem ; 79: 117150, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36640594

RESUMO

N-Methyl-D-aspartate receptors (NMDARs) are key regulators of synaptic plasticity in the central nervous system. Potentiation of NMDARs containing GluN2A subunit has been recently recognized as a promising therapeutic approach for neurological disorders. We identified a novel series of GluN2A positive allosteric modulator (PAM) with a pyridin-2-one scaffold. Initial lead compound 1 was discovered through in silico-based screening of virtual ligands with various monocyclic scaffolds. GluN2A PAM activity was increased by introduction of a methyl group at the 6-position of the pyridin-2-one ring and a cyano group in the side chain. Modification of the aromatic ring led to the identification of potent and brain-penetrant 6-methylpyridin-2-one 17 with a negligible binding activity for α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs). Oral administration of 17 significantly enhanced rat hippocampal long-term potentiation (LTP). Thus, 17 would be a useful in vivo pharmacological tool to investigate complex NMDAR functions for the discovery of therapeutics toward diseases associated with NMDAR dysfunction.


Assuntos
Disfunção Cognitiva , Receptores de N-Metil-D-Aspartato , Ratos , Animais , Receptores de N-Metil-D-Aspartato/metabolismo , Hipocampo/metabolismo , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/metabolismo
7.
Bioorg Med Chem ; 56: 116576, 2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35051811

RESUMO

N-Methyl-d-aspartate receptors (NMDARs) are members of the ionotropic glutamate receptor family and play a crucial role in learning and memory by regulating synaptic plasticity. Activation of NMDARs containing GluN2A, one of the NMDAR subunits, has recently attracted attention as a promising therapeutic approach for neuropsychiatric diseases such as schizophrenia, depression, and epilepsy. In the present study, we developed potent and brain-penetrable GluN2A-selective positive allosteric modulators. Lead compound 2b was generated by scaffold hopping of hit compound 1, identified from the internal alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR)-focused compound library through a high-throughput screening campaign. Subsequent optimization of the lead compound, including a structure-based drug design approach, resulted in the identification of a potent GluN2A PAM (R)-9, which possessed high selectivity against both subtypes of AMPAR and NMDAR. Furthermore, (R)-9 significantly enhanced long-term potentiation in the rat hippocampus 24 h after oral administration, indicating that this molecule is a potentially useful in vivo pharmacological tool for treating psychiatric diseases.


Assuntos
Encéfalo/metabolismo , Descoberta de Drogas , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Administração Oral , Regulação Alostérica/efeitos dos fármacos , Animais , Sítios de Ligação/efeitos dos fármacos , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Injeções Intravenosas , Masculino , Simulação de Acoplamento Molecular , Estrutura Molecular , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade
8.
Int J Mol Sci ; 23(16)2022 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-36012124

RESUMO

NX210c is a disease-modifying dodecapeptide derived from the subcommissural organ-spondin that is under preclinical and clinical development for the treatment of neurological disorders. Here, using whole-cell patch-clamp recordings, we demonstrate that NX210c increased α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR)- and GluN2A-containing N-methyl-D-aspartate receptor (GluN2A-NMDAR)-mediated excitatory postsynaptic currents in the brain. Accordingly, using extracellular field excitatory postsynaptic potential recordings, an enhancement of synaptic transmission was shown in the presence of NX210c in two different neuronal circuits. Furthermore, the modulation of synaptic transmission and GluN2A-NMDAR-driven signaling by NX210c restored memory in mice chronically treated with the NMDAR antagonist phencyclidine. Overall, by promoting glutamatergic receptor-related neurotransmission and signaling, NX210c represents an innovative therapeutic opportunity for patients suffering from CNS disorders, injuries, and states with crippling synaptic dysfunctions.


Assuntos
Receptores de AMPA , Transmissão Sináptica , Animais , Sistema Nervoso Central/metabolismo , Potenciais Pós-Sinápticos Excitadores/fisiologia , Camundongos , Peptídeos , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transmissão Sináptica/fisiologia
9.
J Neurosci ; 40(50): 9564-9575, 2020 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-33158959

RESUMO

d-serine is the primary NMDAR coagonist at mature forebrain synapses and is synthesized by the enzyme serine racemase (SR). However, our understanding of the mechanisms regulating the availability of synaptic d-serine remains limited. Though early studies suggested d-serine is synthesized and released from astrocytes, more recent studies have demonstrated a predominantly neuronal localization of SR. More specifically, recent work intriguingly suggests that SR may be found at the postsynaptic density, yet the functional implications of postsynaptic SR on synaptic transmission are not yet known. Here, we show an age-dependent dendritic and postsynaptic localization of SR and d-serine by immunohistochemistry and electron microscopy in mouse CA1 pyramidal neurons. In addition, using a single-neuron genetic approach in SR conditional KO mice from both sexes, we demonstrate a cell-autonomous role for SR in regulating synaptic NMDAR function at Schaffer collateral (CA3)-CA1 synapses. Importantly, single-neuron genetic deletion of SR resulted in the elimination of LTP at 1 month of age, which could be rescued by exogenous d-serine. Interestingly, there was a restoration of LTP by 2 months of age that was associated with an upregulation of synaptic GluN2B. Our findings support a cell-autonomous role for postsynaptic neuronal SR in regulating synaptic NMDAR function and suggests a possible autocrine mode of d-serine action.SIGNIFICANCE STATEMENT NMDARs are key regulators of neurodevelopment and synaptic plasticity and are unique in their requirement for binding of a coagonist, which is d-serine at most forebrain synapses. However, our understanding of the mechanisms regulating synaptic d-serine availability remains limited. d-serine is synthesized in the brain by the neuronal enzyme serine racemase (SR). Here, we show dendritic and postsynaptic localization of SR and d-serine in CA1 pyramidal neurons. In addition, using single-neuron genetic deletion of SR, we establish a role of postsynaptic SR in regulating NMDAR function. These results support an autocrine mode of d-serine action at synapses.


Assuntos
Dendritos/metabolismo , Células Piramidais/metabolismo , Racemases e Epimerases/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/metabolismo , Fatores Etários , Animais , Região CA1 Hipocampal/metabolismo , Feminino , Masculino , Camundongos , Camundongos Knockout , Plasticidade Neuronal/fisiologia , Racemases e Epimerases/genética , Transmissão Sináptica/fisiologia
10.
J Biol Chem ; 295(50): 17281-17297, 2020 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-33037073

RESUMO

The adipocyte-derived hormone leptin increases trafficking of KATP and Kv2.1 channels to the pancreatic ß-cell surface, resulting in membrane hyperpolarization and suppression of insulin secretion. We have previously shown that this effect of leptin is mediated by the NMDA subtype of glutamate receptors (NMDARs). It does so by potentiating NMDAR activity, thus enhancing Ca2+ influx and the ensuing downstream signaling events that drive channel trafficking to the cell surface. However, the molecular mechanism by which leptin potentiates NMDARs in ß-cells remains unknown. Here, we report that leptin augments NMDAR function via Src kinase-mediated phosphorylation of the GluN2A subunit. Leptin-induced membrane hyperpolarization diminished upon pharmacological inhibition of GluN2A but not GluN2B, indicating involvement of GluN2A-containing NMDARs. GluN2A harbors tyrosine residues that, when phosphorylated by Src family kinases, potentiate NMDAR activity. We found that leptin increases phosphorylation of Tyr-418 in Src, an indicator of kinase activation. Pharmacological inhibition of Src or overexpression of a kinase-dead Src mutant prevented the effect of leptin, whereas a Src kinase activator peptide mimicked it. Using mutant GluN2A overexpression, we show that Tyr-1292 and Tyr-1387 but not Tyr-1325 are responsible for the effect of leptin. Importantly, ß-cells from db/db mice, a type 2 diabetes mouse model lacking functional leptin receptors, or from obese diabetic human donors failed to respond to leptin but hyperpolarized in response to NMDA. Our study reveals a signaling pathway wherein leptin modulates NMDARs via Src to regulate ß-cell excitability and suggests NMDARs as a potential target to overcome leptin resistance.


Assuntos
Células Secretoras de Insulina/metabolismo , Leptina/metabolismo , Potenciais da Membrana , Receptores de N-Metil-D-Aspartato/metabolismo , Quinases da Família src/metabolismo , Animais , Linhagem Celular , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Leptina/genética , Camundongos , Camundongos Mutantes , Mutação , Obesidade/genética , Obesidade/metabolismo , Fosforilação , Receptores de N-Metil-D-Aspartato/genética , Quinases da Família src/genética
11.
Eur J Neurosci ; 54(2): 4456-4474, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34048091

RESUMO

The NMDA receptors (NMDARs) expressed by AII and A17 amacrine cells, the two main inhibitory interneurons of the rod pathway microcircuit in the mammalian retina, are exclusively extrasynaptic, activated by ambient levels of glutamate, and molecularly distinct, with AII and A17 amacrines expressing GluN2B- and GluN2A-containing receptors, respectively. This important sensory microcircuit thus provides a unique model to study the activation and function of extrasynaptic NMDARs. Here, we investigated the sources of glutamate and the endogenous co-agonists (d-serine or glycine) that activate these distinct populations of NMDARs. With acute slices from rat retina, we used whole-cell voltage-clamp recording and measurement of current noise to monitor levels of NMDAR activity. Pre-incubation of retina with bafilomycin A1 (an inhibitor of neurotransmitter uptake into synaptic vesicles) abolished NMDAR-mediated noise in AII, but not A17 amacrines, suggesting a vesicular source of glutamate activates AII NMDARs, whereas a non-vesicular source activates A17 NMDARs. Pre-incubation of retina with l-methionine sulfoximine (an inhibitor of glutamine synthetase) also abolished NMDAR-mediated noise in AII, but not A17 amacrines, suggesting a neuronal source of glutamate activates AII NMDARs, whereas a glial source activates A17 NMDARs. Enzymatic breakdown of d-serine reduced NMDAR-mediated noise in AII, but not A17 amacrines, suggesting d-serine is the endogenous co-agonist at AII, but not A17 NMDARs. Our results reveal unique characteristics of these two populations of extrasynaptic NMDARs. The differential and independent activation of these receptors is likely to provide specific contributions to the signal processing and plasticity of the cellular components of the rod pathway microcircuit.


Assuntos
Células Amácrinas , Receptores de N-Metil-D-Aspartato , Animais , Ácido Glutâmico , Técnicas de Patch-Clamp , Ratos , Retina , Sinapses
12.
Exp Brain Res ; 239(9): 2701-2709, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34223957

RESUMO

To investigate the role of GluN2A and GluN2B in neuroprotective effect of sevoflurane preconditioning against cerebral ischemia-reperfusion injury (CIRI). Rats were randomly divided into five groups as follows: control, ischemia-reperfusion (I/R) 6 h, sevoflurane preconditioning (SP), SP + amantadine, SP + NMDA. Immunoblot and immunoprecipitation were used to detect the tyrosine phosphorylation of GluN2A/GluN2B, the interaction of GluN2A/GluN2B-PSD-95-MLK3 and the expression of phosphorylation of MLK3, MKK7 and JNK3. Cresyl violet staining was employed to analyse neuronal injury in rat hippocampal CA1 subfields. Sevoflurane preconditioning inhibits the tyrosine phosphorylation of GluN2A/GluN2B, the interaction of GluN2A/GluN2B-PSD-95-MLK3 and the phosphorylation of MLK3, MKK7 and JNK3 in rat hippocampus. An N-methyl-D-aspartate receptor (NMDAR) antagonist amantadine reversed the MLK3-MKK7- JNK3 signal events. Such reversion was also realized by NMDA (60 and 80 nmol) and low doses of NMDA (0-40 nmol) could not change the inhibitory effect of sevoflurane preconditioning on MLK3-MKK7-JNK3 signal events. Finally, Cresyl violet staining also confirmed that low dose of NMDA reduced neuronal loss in rat hippocampal CA1 subfields. Sevoflurane preconditioning provides neuroprotection against CIRI by inhibiting NMDAR over-activation.


Assuntos
Lesões Encefálicas , Traumatismo por Reperfusão , Animais , MAP Quinase Quinase Quinases/metabolismo , Neuroproteção , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato , Reperfusão , Traumatismo por Reperfusão/tratamento farmacológico , Sevoflurano
13.
Addict Biol ; 26(2): e12939, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32720424

RESUMO

Ethanol consumption impairs learning and memory through disturbances of NMDA-type glutamate receptor-dependent synaptic plasticity (long-term depression [LTD] and long-term potentiation [LTP]) in the hippocampus. Recently, we demonstrated that two ethanol binge-like episodes in young adult rats selectively blocked NMDA-LTD in hippocampal slices, increased NMDA receptor sensitivity to a GluN2B subunit antagonist, and induced cognitive deficits. Here, using knockout adult mice, we show that a stress-responsive transcription factor of the heat shock factor family, HSF2, which is involved in the perturbation of brain development induced by ethanol, participates in these processes. In the absence of ethanol, hsf2-/- mice show a selective loss of LTD in the hippocampus, which is associated with an increased sensitivity of NMDA-field excitatory postsynaptic potentials (fEPSPs) to a GluN2B antagonist, compared with wild-type (WT) mice. These results suggest that HSF2 is required for proper glutamatergic synaptic transmission and LTD plasticity. After 1 month of chronic ethanol consumption in a two-bottle choice paradigm, WT mice showed an increase in hippocampal synaptic transmission, an enhanced sensitivity to GluN2B antagonist, and a blockade of LTD. In contrast, such modulation of synaptic transmission and plasticity were absent in hsf2-/- mice. We conclude that HSF2 is an important mediator of both glutamatergic neurotransmission and synaptic plasticity in basal conditions and also mediates ethanol-induced neuroadaptations of the hippocampus network after chronic ethanol intake.


Assuntos
Etanol/farmacologia , Fatores de Transcrição de Choque Térmico/efeitos dos fármacos , Potenciação de Longa Duração/efeitos dos fármacos , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , N-Metilaspartato/efeitos dos fármacos , Adolescente , Adulto , Fatores Etários , Animais , Hipocampo/efeitos dos fármacos , Humanos , Camundongos
14.
J Neurosci ; 39(4): 627-650, 2019 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-30459218

RESUMO

In the rod pathway of the mammalian retina, axon terminals of glutamatergic rod bipolar cells are presynaptic to AII and A17 amacrine cells in the inner plexiform layer. Recent evidence suggests that both amacrines express NMDA receptors, raising questions concerning molecular composition, localization, activation, and function of these receptors. Using dual patch-clamp recording from synaptically connected rod bipolar and AII or A17 amacrine cells in retinal slices from female rats, we found no evidence that NMDA receptors contribute to postsynaptic currents evoked in either amacrine. Instead, NMDA receptors on both amacrine cells were activated by ambient glutamate, and blocking glutamate uptake increased their level of activation. NMDA receptor activation also increased the frequency of GABAergic postsynaptic currents in rod bipolar cells, suggesting that NMDA receptors can drive release of GABA from A17 amacrines. A striking dichotomy was revealed by pharmacological and immunolabeling experiments, which found GluN2B-containing NMDA receptors on AII amacrines and GluN2A-containing NMDA receptors on A17 amacrines. Immunolabeling also revealed a clustered organization of NMDA receptors on both amacrines and a close spatial association between GluN2B subunits and connexin 36 on AII amacrines, suggesting that NMDA receptor modulation of gap junction coupling between these cells involves the GluN2B subunit. Using multiphoton Ca2+ imaging, we verified that activation of NMDA receptors evoked an increase of intracellular Ca2+ in dendrites of both amacrines. Our results suggest that AII and A17 amacrines express clustered, extrasynaptic NMDA receptors, with different and complementary subunits that are likely to contribute differentially to signal processing and plasticity.SIGNIFICANCE STATEMENT Glutamate is the most important excitatory neurotransmitter in the CNS, but not all glutamate receptors transmit fast excitatory signals at synapses. NMDA-type glutamate receptors act as voltage- and ligand-gated ion channels, with functional properties determined by their specific subunit composition. These receptors can be found at both synaptic and extrasynaptic sites on neurons, but the role of extrasynaptic NMDA receptors is unclear. Here, we demonstrate that retinal AII and A17 amacrine cells, postsynaptic partners at rod bipolar dyad synapses, express extrasynaptic (but not synaptic) NMDA receptors, with different and complementary GluN2 subunits. The localization of GluN2A-containing receptors to A17s and GluN2B-containing receptors to AIIs suggests a mechanism for differential modulation of excitability and signaling in this retinal microcircuit.


Assuntos
Células Amácrinas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Células Amácrinas/efeitos dos fármacos , Células Amácrinas/ultraestrutura , Animais , Cálcio/metabolismo , Conexinas/metabolismo , Dendritos/metabolismo , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Feminino , Junções Comunicantes/efeitos dos fármacos , Técnicas In Vitro , Técnicas de Patch-Clamp , Ratos , Ratos Wistar , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Células Bipolares da Retina/efeitos dos fármacos , Células Bipolares da Retina/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/ultraestrutura , Transdução de Sinais/efeitos dos fármacos , Ácido gama-Aminobutírico/fisiologia , Proteína delta-2 de Junções Comunicantes
15.
J Biol Chem ; 294(29): 11154-11165, 2019 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-31167782

RESUMO

Homocysteine, a metabolite of the methionine cycle, is a known agonist of N-methyl-d-aspartate receptor (NMDAR), a glutamate receptor subtype and is involved in NMDAR-mediated neurotoxicity. Our previous findings have shown that homocysteine-induced, NMDAR-mediated neurotoxicity is facilitated by a sustained increase in phosphorylation and activation of extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK MAPK). In the current study, we investigated the role GluN1/GluN2A-containing functional NMDAR (GluN2A-NMDAR) and GluN1/GluN2B-containing functional NMDAR (GluN2B-NMDAR) in homocysteine-induced neurotoxicity. Our findings revealed that exposing primary cortical neuronal cultures to homocysteine leads to a sustained low-level increase in intracellular Ca2+ We also showed that pharmacological inhibition of GluN2A-NMDAR or genetic deletion of the GluN2A subunit attenuates homocysteine-induced increase in intracellular Ca2+ Our results further established the role of GluN2A-NMDAR in homocysteine-mediated sustained ERK MAPK phosphorylation and neuronal cell death. Of note, the preferential role of GluN2A-NMDAR in homocysteine-induced neurotoxicity was distinctly different from glutamate-NMDAR-induced excitotoxic cell death that involves overactivation of GluN2B-NMDAR and is independent of ERK MAPK activation. These findings indicate a critical role of GluN2A-NMDAR-mediated signaling in homocysteine-induced neurotoxicity.


Assuntos
Cálcio/metabolismo , Morte Celular/efeitos dos fármacos , Homocisteína/farmacologia , Neurônios/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Ativação Enzimática , Feminino , Transporte de Íons , Camundongos , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neurônios/citologia , Fosforilação , Gravidez , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/genética
16.
Neurobiol Dis ; 140: 104850, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32247039

RESUMO

Mutations in synaptic NMDA receptors (NMDARs) are associated with epilepsy and neurodevelopmental disorders. The effects of several such mutations have been investigated in recombinantly-expressed NMDARs under conditions of steady-state activation. Such experiments provide only limited insight into how mutations affect NMDAR-mediated excitatory synaptic currents (EPSCs). The present study aimed to characterize the effects of the GluN2AN615K, GluN2BN615I and GluN2BV618G gain-of-function mutations on EPSCs mediated by diheteromeric GluN1/2A and GluN1/2B receptors and triheteromeric GluN1/2A/2B receptors, as these are the most abundant synaptic NMDARs in vivo. Subunit composition was controlled by studying 'artificial' synapses formed between cultured neurons (which provide presynaptic terminals) and HEK293 cells that express the NMDAR subunits of interest plus the synapse-promoting molecule, neuroligin-1B. When incorporated into diheteromeric receptors, all three mutations ablated voltage-dependent Mg2+ block of EPSCs, as previously shown. In addition, we were surprised to find that increasing external Mg2+ from 0 to 1 mM strongly enhanced the magnitude of EPSCs mediated by mutant diheteromers. In contrast, triheteromeric receptors exhibited normal voltage-dependent Mg2+ block. The GluN2AN615K mutation also slowed the decay of GluN1/2A/2B- but not GluN1/2A-mediated EPSCs. The GluN2BN615I mutation enhanced the magnitude of both GluN1/2B- and GluN1/2A/2B-mediated EPSCs. The GluN2BV618G mutation enhanced the magnitude of both GluN1/2B- and GluN1/2A/2B-mediated EPSCs, although these effects were partly compensated by a faster EPSC decay rate. The mutations also diminished the potency of the anti-epileptic pore-blocker, memantine, thus explaining the lack of memantine efficacy in patients with GluN2BN615I or GluN2BV618G mutations. Given these effects, the three mutations would be expected to enhance the cation influx rate and thereby contribute to epilepsy phenotypes.


Assuntos
Epilepsia/genética , Mutação com Ganho de Função , Receptores de N-Metil-D-Aspartato/genética , Sinapses/fisiologia , Animais , Feminino , Células HEK293 , Humanos , Masculino , Neurônios/fisiologia , Técnicas de Patch-Clamp , Ratos
17.
Biochem Biophys Res Commun ; 522(4): 1046-1051, 2020 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-31818458

RESUMO

N-methyl-d-aspartate receptor (NMDAR) is an essential target for ethanol action in the central nervous system (CNS). Whereas an alcohol addiction treatment represents a severe medical problem, many aspects of ethanol action at physiologically relevant concentrations on NMDARs are still unclear. Here using the whole-cell patch-clamp recording on cortical neurons in the primary culture, we studied inhibition of NMDAR currents by different ethanol concentrations ([Et]s) and its dependence on extracellular Ca2+. The ethanol action on NMDA-activated currents exhibited a biphasic concentration-inhibition relationship in the presence of extracellular Ca2+. The high-affinity region of the curve was found within the range of [Et]s from 9 mM to 30 mM and was characterized by IC50,H of about 20 mM. The low-affinity region was observed within the range of [Et]s from 85 mM to 200 mM with IC50,L of about 150 mM. In the absence of extracellular Ca2+, the ethanol concentration-inhibition relationship became monophasic, with IC50,L of about 200 mM, since the high-affinity component disappeared. A substitution of Li+ for Na+ in the bathing solution and an extraction of cholesterol from the plasma membrane with methyl-ß-cyclodextrin, which are the treatments that both promote the Ca2+-dependent desensitization (CDD) of NMDARs, abolished the high-affinity Ca2+-dependent component of the NMDAR ethanol inhibition. Besides, this component was not observed when neurons were loaded with BAPTA. These data suggest that most likely, ethanol at low concentrations enhances the NMDAR CDD. In agreement when the dependence of the NMDAR CDD on extracellular Ca2+ was directly measured 22 mM ethanol enhanced the NMDAR CDD since an extracellular Ca2+ concentration that caused 50% of the NMDAR CDD decreased almost 3-folds from 0.81 mM to 0.28 mM, and an extent of the CDD was also more pronounced. The low-affinity component of the NMDAR ethanol inhibition was resistant to the above treatments suggesting CDD-independent direct action on NMDARs. Thus, at a physiologically relevant concentration of extracellular Ca2+ and ethanol that could be reached in the blood during light-mild human alcohol intoxication, ethanol causes an enhancement of the NMDAR CDD, which could be in general accompanied by some disruptions of the CNS excitatory system.


Assuntos
Cálcio/metabolismo , Etanol/toxicidade , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Espaço Extracelular/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Lítio/farmacologia , N-Metilaspartato/farmacologia , Ratos Wistar , beta-Ciclodextrinas
18.
Neurobiol Learn Mem ; 171: 107210, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32145408

RESUMO

Epigenetic mechanisms of learning and memory are particularly interesting topics in neuroscience that have recently been investigated. As shown in our previous study, IQGAP1, a scaffolding protein of MAPK, is involved in fear memory through interactions with GluN2A-containing NMDA receptors and the ERK1/2 cascade. However, researchers have not determined whether histone posttranslational modifications are regulated by the IQGAP1/ERK signaling pathway. We performed in vivo studies using IQGAP1-/- and IQGAP1+/+ mice to provide insights into the specific functions of IQGAP1 in memory processes and the precise mechanisms underlying its regulatory effects. IQGAP1-/- mice exhibited impaired fear memory, decreased levels of phosphorylated ERK1/2 and histone H3S10, decreased acetylation of H3K14, and decreased c-Fos expression in the hippocampus compared to IQGAP1+/+ mice after fear conditioning. HDAC2 was significantly enriched at the c-fos gene promoter in IQGAP1-/- mice. Correspondingly, the disruption of the epigenetic regulation induced by ERK1/2 signaling through an intra-hippocampal injection of the MEK antagonist U0126 or GluN2A-selective pharmacological antagonist NVP-AAM077 blocked context-dependent memory formation, while no changes were observed after treatment with the GluN2B-selective antagonist Ro25-6981. The administration of SAHA, a non-specific HDAC inhibitor, or knock-down of HDAC2 with shHDAC2-AAV in the dorsal hippocampus significantly rescued the impaired fear memory formation, H3S10 phosphorylation, H3K14 acetylation, and c-Fos expression in IQGAP1-/- mice. Thus, we postulated that the IQGAP1/ERK-dependent mechanism regulating histone posttranslational modifications via HDAC2 potentially underlies memory formation.


Assuntos
Medo/fisiologia , Histona Desacetilase 2/metabolismo , Memória/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Proteínas Ativadoras de ras GTPase/metabolismo , Animais , Butadienos/farmacologia , Inibidores Enzimáticos/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Medo/efeitos dos fármacos , Histona Desacetilase 2/genética , Histonas/metabolismo , Memória/efeitos dos fármacos , Camundongos , Camundongos Knockout , Nitrilas/farmacologia , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Quinoxalinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteínas Ativadoras de ras GTPase/genética
19.
Arch Biochem Biophys ; 685: 108348, 2020 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-32198047

RESUMO

Memory formation transpires to be by activation and persistent modification of synapses. A chain of biochemical events accompany synaptic activation and culminate in memory formation. These biochemical events are steered by interplay and modulation of various synaptic proteins, achieved by conformational changes and phosphorylation/dephosphorylation of these proteins. Calcium/calmodulin dependent protein kinase II (CaMKII) and N-methyl-d-aspartate receptors (NMDARs) are synaptic proteins whose interactions play a pivotal role in learning and memory process. Catalytic activity of CaMKII is modulated upon its interaction with the GluN2B subunit of NMDAR. The structural basis of this interaction is not clearly understood. We have investigated the role of Glu60 of α-CaMKII, a conserved residue present in the ATP binding region of kinases, in the regulation of catalysis of CaMKII by GluN2B. Mutation of Glu60 to Gly exerts different effects on the kinetic parameters of phosphorylation of GluN2B and GluN2A, of which only GluN2B binds to the T-site of CaMKII. GluN2B induced modulation of the kinetic parameters of peptide substrate was altered in the E60G mutant. The mutation almost abolished the modulation of the apparent Km value for protein substrate. However, although kinetic parameters for ATP were altered by mutating Glu60, modulation of the apparent Km value for ATP by GluN2B seen in WT was exhibited by the E60G mutant of α-CaMKII. Hence our results posit that the communication of the T-site of CaMKII with protein substrate binding region of active site is mediated through Glu60 while the communication of the T-site with the ATP binding region is not entirely dependent on Glu60.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Ácido Glutâmico/química , Receptores de N-Metil-D-Aspartato/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/química , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Calmodulina/metabolismo , Domínio Catalítico , Células HEK293 , Humanos , Cinética , Mutação , Ligação Proteica
20.
Alcohol Clin Exp Res ; 44(2): 479-491, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31872888

RESUMO

BACKGROUND: N-methyl-D-aspartate receptors (NMDARs) are glutamate-activated, heterotetrameric ligand-gated ion channels critically important in virtually all aspects of glutamatergic signaling. Ethanol (EtOH) inhibition of NMDARs is thought to mediate specific actions of EtOH during acute and chronic exposure. Studies from our laboratory, and others, identified EtOH-sensitive sites within specific transmembrane (TM) domains involved in channel gating as well as those in subdomains of extracellular and intracellular regions of GluN1 and GluN2 subunits that affect channel function. In this study, we characterize for the first time the physiological and behavioral effects of EtOH on knock-in mice expressing a GluN2A subunit that shows reduced sensitivity to EtOH. METHODS: A battery of tests evaluating locomotion, anxiety, sedation, motor coordination, and voluntary alcohol intake were performed in wild-type mice and those expressing the GluN2A A825W knock-in mutation. Whole-cell patch-clamp electrophysiological recordings were used to confirm reduced EtOH sensitivity of NMDAR-mediated currents in 2 separate brain regions (mPFC and the cerebellum) where the GluN2A subunit is known to contribute to NMDAR-mediated responses. RESULTS: Male and female mice homozygous for the GluN2A(A825W) knock-in mutation showed reduced EtOH inhibition of NMDAR-mediated synaptic currents in mPFC and cerebellar neurons as compared to their wild-type counterparts. GluN2A(A825W) male but not female mice were less sensitive to the sedative and motor-incoordinating effects of EtOH and showed a rightward shift in locomotor-stimulating effects of EtOH. There was no effect of the mutation on EtOH-induced anxiolysis or voluntary EtOH consumption in either male or female mice. CONCLUSIONS: These findings show that expression of EtOH-resistant GluN2A NMDARs results in selective and sex-specific changes in the behavioral sensitivity to EtOH.


Assuntos
Consumo de Bebidas Alcoólicas/genética , Consumo de Bebidas Alcoólicas/metabolismo , Etanol/administração & dosagem , Técnicas de Introdução de Genes/métodos , Receptores de N-Metil-D-Aspartato/biossíntese , Receptores de N-Metil-D-Aspartato/genética , Animais , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Feminino , Expressão Gênica , Locomoção/efeitos dos fármacos , Locomoção/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA