Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 6: 36684, 2016 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-27857196

RESUMO

N-methyl-D-aspartate receptors (NMDARs) are necessary for the induction of synaptic plasticity and for the consolidation of learning and memory. NMDAR function is tightly regulated by functionally opposed families of kinases and phosphatases. Herein we show that the striatal-enriched protein tyrosine phosphatase (STEP) is recruited by Gαq-coupled receptors, including the M1 muscarinic acetylcholine receptor (M1R), and opposes the Src tyrosine kinase-mediated increase in the function of NMDARs composed of GluN2A. STEP activation by M1R stimulation requires IP3Rs and can depress NMDA-evoked currents with modest intracellular Ca2+ buffering. Src recruitment by M1R stimulation requires coincident NMDAR activation and can augment NMDA-evoked currents with high intracellular Ca2+ buffering. Our findings suggest that Src and STEP recruitment is contingent on differing intracellular Ca2+ dynamics that dictate whether NMDAR function is augmented or depressed following M1R stimulation.


Assuntos
Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Quinases da Família src/metabolismo , Animais , Animais Geneticamente Modificados , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/metabolismo , Plasticidade Neuronal , Proteínas Tirosina Fosfatases não Receptoras/genética , Ratos , Ratos Wistar , Receptor Muscarínico M1/metabolismo
2.
J Neurosci ; 35(45): 15157-69, 2015 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-26558786

RESUMO

In Alzheimer's disease, accumulation of soluble oligomers of ß-amyloid peptide is known to be highly toxic, causing disturbances in synaptic activity and neuronal death. Multiple studies relate these effects to increased oxidative stress and aberrant activity of calcium-permeable cation channels leading to calcium imbalance. The transient receptor potential melastatin 2 (TRPM2) channel, a Ca(2+)-permeable nonselective cation channel activated by oxidative stress, has been implicated in neurodegenerative diseases, and more recently in amyloid-induced toxicity. Here we show that the function of TRPM2 is augmented by treatment of cultured neurons with ß-amyloid oligomers. Aged APP/PS1 Alzheimer's mouse model showed increased levels of endoplasmic reticulum stress markers, protein disulfide isomerase and phosphorylated eukaryotic initiation factor 2α, as well as decreased levels of the presynaptic marker synaptophysin. Elimination of TRPM2 in APP/PS1 mice corrected these abnormal responses without affecting plaque burden. These effects of TRPM2 seem to be selective for ß-amyloid toxicity, as ER stress responses to thapsigargin or tunicamycin in TRPM2(-/-) neurons was identical to that of wild-type neurons. Moreover, reduced microglial activation was observed in TRPM2(-/-)/APP/PS1 hippocampus compared with APP/PS1 mice. In addition, age-dependent spatial memory deficits in APP/PS1 mice were reversed in TRPM2(-/-)/APP/PS1 mice. These results reveal the importance of TRPM2 for ß-amyloid neuronal toxicity, suggesting that TRPM2 activity could be potentially targeted to improve outcomes in Alzheimer's disease. SIGNIFICANCE STATEMENT: Transient receptor potential melastatin 2 (TRPM2) is an oxidative stress sensing calcium-permeable channel that is thought to contribute to calcium dysregulation associated with neurodegenerative diseases, including Alzheimer's disease. Here we show that oligomeric ß-amyloid, the toxic peptide in Alzheimer's disease, facilitates TRPM2 channel activation. In mice designed to model Alzheimer's disease, genetic elimination of TRPM2 normalized deficits in synaptic markers in aged mice. Moreover, the absence of TRPM2 improved age-dependent spatial memory deficits observed in Alzheimer's mice. Our results reveal the importance of TRPM2 for neuronal toxicity and memory impairments in an Alzheimer's mouse model and suggest that TRPM2 could be targeted for the development of therapeutic agents effective in the treatment of dementia.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Transtornos da Memória/metabolismo , Canais de Cátion TRPM/deficiência , Doença de Alzheimer/induzido quimicamente , Peptídeos beta-Amiloides/toxicidade , Animais , Células Cultivadas , Humanos , Masculino , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Placa Amiloide/metabolismo , Canais de Cátion TRPM/genética
3.
Hippocampus ; 24(12): 1601-14, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25087967

RESUMO

Endocannabinoids (eCBs), including AEA and 2-AG, are endogenous signaling mediators involved in many physiological and pathological events. The G protein-coupled cannabinoid receptor 1 (CB1 R) is an important target for eCBs, however, additional non-CB1 receptor targets have also been identified. Although recent evidence suggests that NMDA receptor function may be regulated by eCBs, the underlying mechanisms remain poorly characterized. Using acutely isolated CA1 neurons and slices from the hippocampus, we found that both AEA and 2-AG potentiate NMDAR-mediated currents independently of CB1 receptors (CB1 Rs) and via distinct signaling pathways. Potentiation by AEA requires the activation of TRPV1 channels. In contrast, potentiation by 2-AG requires the sequential activation of PKC and Src. Additionally, in hippocampal slices, we found that both AEA and 2-AG induce NMDAR-mediated metaplasticity and facilitate the induction of subsequent LTD independently of CB1 Rs. Enhanced LTD by AEA, but not 2-AG, was dependent on TRPV1 channels. Our findings reveal previously unrecognized non-CB1 R-dependent signaling cascades through which the two major eCBs regulate NMDA receptor function and consequently synaptic plasticity.


Assuntos
Ácidos Araquidônicos/metabolismo , Região CA1 Hipocampal/fisiologia , Endocanabinoides/metabolismo , Glicerídeos/metabolismo , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Alcamidas Poli-Insaturadas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Região CA1 Hipocampal/efeitos dos fármacos , Cálcio/metabolismo , Células Cultivadas , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Masculino , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Ratos Wistar , Receptor CB1 de Canabinoide/metabolismo , Canais de Cátion TRPV/metabolismo , Técnicas de Cultura de Tecidos
4.
Mol Brain ; 7: 15, 2014 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-24594013

RESUMO

BACKGROUND: Excess expression of acetylcholinesterase (AChE) in the cortex and hippocampus causes a decrease in the number of glutamatergic synapses and alters the expression of neurexin and neuroligin, trans-synaptic proteins that control synaptic stability. The molecular sequence and three-dimensional structure of AChE are homologous to the corresponding aspects of the ectodomain of neuroligin. This study investigated whether excess AChE interacts physically with neurexin to destabilize glutamatergic synapses. RESULTS: The results showed that AChE clusters colocalized with neurexin assemblies in the neurites of hippocampal neurons and that AChE co-immunoprecipitated with neurexin from the lysate of these neurons. Moreover, when expressed in human embryonic kidney 293 cells, N-glycosylated AChE co-immunoprecipitated with non-O-glycosylated neurexin-1ß, with N-glycosylation of the AChE being required for this co-precipitation to occur. Increasing extracellular AChE decreased the association of neurexin with neuroligin and inhibited neuroligin-induced synaptogenesis. The number and activity of excitatory synapses in cultured hippocampal neurons were reduced by extracellular catalytically inactive AChE. CONCLUSIONS: Excessive glycosylated AChE could competitively disrupt a subset of the neurexin-neuroligin junctions consequently impairing the integrity of glutamatergic synapses. This might serve a molecular mechanism of excessive AChE induced neurodegeneration.


Assuntos
Acetilcolinesterase/metabolismo , Glutamatos/metabolismo , Hipocampo/citologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Sinapses/metabolismo , Animais , Moléculas de Adesão Celular Neuronais/metabolismo , Membrana Celular/metabolismo , Glicosilação , Células HEK293 , Humanos , Imunoprecipitação , Ligantes , Modelos Biológicos , Ligação Proteica , Splicing de RNA , Ratos , Ratos Wistar
5.
Int J Mol Sci ; 15(2): 3003-24, 2014 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-24562329

RESUMO

G Protein Coupled Receptors (GPCRs) are the largest family of receptors whose ligands constitute nearly a third of prescription drugs in the market. They are widely involved in diverse physiological functions including learning and memory. NMDA receptors (NMDARs), which belong to the ionotropic glutamate receptor family, are likewise ubiquitously expressed in the central nervous system (CNS) and play a pivotal role in learning and memory. Despite its critical contribution to physiological and pathophysiological processes, few pharmacological interventions aimed directly at regulating NMDAR function have been developed to date. However, it is well established that NMDAR function is precisely regulated by cellular signalling cascades recruited downstream of G protein coupled receptor (GPCR) stimulation. Accordingly, the downstream regulation of NMDARs likely represents an important determinant of outcome following treatment with neuropsychiatric agents that target selected GPCRs. Importantly, the functional consequence of such regulation on NMDAR function varies, based not only on the identity of the GPCR, but also on the cell type in which relevant receptors are expressed. Indeed, the mechanisms responsible for regulating NMDARs by GPCRs involve numerous intracellular signalling molecules and regulatory proteins that vary from one cell type to another. In the present article, we highlight recent findings from studies that have uncovered novel mechanisms by which selected GPCRs regulate NMDAR function and consequently NMDAR-dependent plasticity.


Assuntos
Receptores Acoplados a Proteínas G/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sistema Nervoso Central/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Humanos , Plasticidade Neuronal , Subunidades Proteicas/metabolismo
6.
J Neurosci ; 33(42): 16552-64, 2013 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-24133259

RESUMO

In Alzheimer's disease (AD), soluble amyloid-ß oligomers (AßOs) trigger neurotoxic signaling, at least partially, via the cellular prion protein (PrP(C)). However, it is unknown whether other ligands of PrP(C) can regulate this potentially toxic interaction. Stress-inducible phosphoprotein 1 (STI1), an Hsp90 cochaperone secreted by astrocytes, binds to PrP(C) in the vicinity of the AßO binding site to protect neurons against toxic stimuli. Here, we investigated a potential role of STI1 in AßO toxicity. We confirmed the specific binding of AßOs and STI1 to the PrP and showed that STI1 efficiently inhibited AßO binding to PrP in vitro (IC50 of ∼70 nm) and also decreased AßO binding to cultured mouse primary hippocampal neurons. Treatment with STI1 prevented AßO-induced synaptic loss and neuronal death in mouse cultured neurons and long-term potentiation inhibition in mouse hippocampal slices. Interestingly, STI1-haploinsufficient neurons were more sensitive to AßO-induced cell death and could be rescued by treatment with recombinant STI1. Noteworthy, both AßO binding to PrP(C) and PrP(C)-dependent AßO toxicity were inhibited by TPR2A, the PrP(C)-interacting domain of STI1. Additionally, PrP(C)-STI1 engagement activated α7 nicotinic acetylcholine receptors, which participated in neuroprotection against AßO-induced toxicity. We found an age-dependent upregulation of cortical STI1 in the APPswe/PS1dE9 mouse model of AD and in the brains of AD-affected individuals, suggesting a compensatory response. Our findings reveal a previously unrecognized role of the PrP(C) ligand STI1 in protecting neurons in AD and suggest a novel pathway that may help to offset AßO-induced toxicity.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Proteínas de Choque Térmico/metabolismo , Neurônios/metabolismo , Proteínas PrPC/metabolismo , Doença de Alzheimer/metabolismo , Animais , Astrócitos/metabolismo , Encéfalo/metabolismo , Células Cultivadas , Hipocampo/metabolismo , Camundongos , Ligação Proteica , Transdução de Sinais/fisiologia , Receptor Nicotínico de Acetilcolina alfa7/metabolismo
7.
Acta Pharmacol Sin ; 34(11): 1381-5, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24162508

RESUMO

The intricate and complex interaction between different populations of neurons in the brain has imposed limits on our ability to gain detailed understanding of synaptic transmission and its integration when employing classical electrophysiological approaches. Indeed, electrical field stimulation delivered via traditional microelectrodes does not permit the targeted, precise and selective control of neuronal activity amongst a varied population of neurons and their inputs (eg, cholinergic, dopaminergic or glutamatergic neurons). Recently established optogenetic techniques overcome these limitations allowing precise control of the target neuron populations, which is essential for the elucidation of the neural substrates underlying complex animal behaviors. Indeed, by introducing light-activated channels (ie, microbial opsin genes) into specific neuronal populations, optogenetics enables non-invasive optical control of specific neurons with milliseconds precision. These approaches can readily be applied to freely behaving live animals. Recently there is increased interests in utilizing optogenetics tools to understand synaptic plasticity and learning/memory. Here, we summarize recent progress in applying optogenetics in in the study of synaptic plasticity.


Assuntos
Plasticidade Neuronal/fisiologia , Optogenética/métodos , Sinapses/fisiologia , Animais , Comportamento Animal/fisiologia , Encéfalo/fisiologia , Humanos , Aprendizagem/fisiologia , Memória/fisiologia
8.
Mol Brain ; 6: 24, 2013 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-23672716

RESUMO

BACKGROUND: N-methyl-D-aspartate (NMDA) receptors are regulated by several G protein-coupled receptors (GPCRs) as well as receptor tyrosine kinases. Serotonin (5-HT) type 7 receptors are expressed throughout the brain including the thalamus and hippocampus. Long-term (2-24 h) activation of 5-HT7 receptors promotes the expression of neuroprotective growth factor receptors, including the platelet-derived growth factor (PDGF) ß receptors which can protect neurons against NMDA-induced neurotoxicity. RESULTS: In contrast to long-term activation of 5-HT7 receptors, acute (5 min) treatment of isolated hippocampal neurons with the 5-HT7 receptor agonist 5-carboxamidotryptamine (5-CT) enhances NMDA-evoked peak currents and this increase in peak currents is blocked by the 5-HT7 receptor antagonist, SB 269970. In hippocampal slices, acute 5-HT7 receptor activation increases NR1 NMDA receptor subunit phosphorylation and differentially alters the phosphorylation state of the NR2B and NR2A subunits. NMDA receptor subunit cell surface expression is also differentially altered by 5-HT7 receptor agonists: NR2B cell surface expression is decreased whereas NR1 and NR2A surface expression are not significantly altered. CONCLUSIONS: In contrast to the negative regulatory effects of long-term activation of 5-HT7 receptors on NMDA receptor signaling, acute activation of 5-HT7 receptors promotes NMDA receptor activity. These findings highlight the potential for temporally differential regulation of NMDA receptors by the 5-HT7 receptor.


Assuntos
Hipocampo/citologia , Ativação do Canal Iônico/efeitos dos fármacos , N-Metilaspartato/farmacologia , Neurônios/metabolismo , Subunidades Proteicas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de Serotonina/metabolismo , Amidas/farmacologia , Animais , Modelos Biológicos , Neurônios/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Fosfosserina/metabolismo , Piperazinas/farmacologia , Ratos Wistar , Serotonina/análogos & derivados , Serotonina/farmacologia
9.
Anesthesiology ; 118(5): 1065-75, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23485993

RESUMO

BACKGROUND: Volatile anesthetics act primarily through upregulating the activity of γ-aminobutyric acid type A (GABAA) receptors. They also exhibit antiinflammatory actions in the lung. Rodent alveolar type II (ATII) epithelial cells express GABAA receptors and the inflammatory factor cyclooxygenase-2 (COX-2). The goal of this study was to determine whether human ATII cells also express GABAA receptors and whether volatile anesthetics upregulate GABAA receptor activity, thereby reducing the expression of COX-2 in ATII cells. METHODS: The expression of GABAA receptor subunits and COX-2 in ATII cells of human lung tissue and in the human ATII cell line A549 was studied with immunostaining and immunoblot analyses. Patch clamp recordings were used to study the functional and pharmacological properties of GABAA receptors in cultured A549 cells. RESULTS: ATII cells in human lungs and cultured A549 cells expressed GABAA receptor subunits and COX-2. GABA induced currents in A549 cells, with half-maximal effective concentration of 2.5 µM. Isoflurane (0.1-250 µM) enhanced the GABA currents, which were partially inhibited by bicuculline. Treating A549 cells with muscimol or with isoflurane (250 µM) reduced the expression of COX-2, an effect that was attenuated by cotreatment with bicuculline. CONCLUSIONS: GABAA receptors expressed by human ATII cells differ pharmacologically from those in neurons, exhibiting a higher affinity for GABA and lower sensitivity to bicuculline. Clinically relevant concentrations of isoflurane increased the activity of GABAA receptors and reduced the expression of COX-2 in ATII cells. These findings reveal a novel mechanism that could contribute to the antiinflammatory effect of isoflurane in the human lung.


Assuntos
Anestésicos Inalatórios/farmacologia , Células Epiteliais/efeitos dos fármacos , Isoflurano/farmacologia , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/efeitos dos fármacos , Receptores de GABA-A/efeitos dos fármacos , Anestésicos Inalatórios/química , Animais , Bicuculina/farmacologia , Western Blotting , Corantes , Ciclo-Oxigenase 2/biossíntese , Imunofluorescência , Agonistas GABAérgicos/farmacologia , Antagonistas GABAérgicos/farmacologia , Humanos , Isoflurano/química , Camundongos , Microscopia Confocal , Muscimol/farmacologia , Técnicas de Patch-Clamp , Soluções , Azul Tripano
10.
Sci Rep ; 3: 926, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23378895

RESUMO

Group II metabotropic glutamate receptors (mGluR2/3) have emerged as important targets for the treatment of schizophrenia. Since hypofunction of N-methyl-D-aspartate receptors (NMDARs) has also been implicated in the etiology of schizophrenia, we examined whether postsynaptic mGluR2/3 regulate NMDAR function. Activation of mGluR2/3 significantly decreased the ratio of AMPA-to-NMDA excitatory postsynaptic currents at Schaffer Collateral-CA1 synapses and enhanced the peak of NMDA-evoked currents in acutely isolated CA1 neurons. The mGluR2/3-mediated potentiation of NMDAR currents was selective for GluN2A-containing NMDARs and was mediated by the Src family kinase Src. Activation of mGluR2/3 inhibited the adenylyl cyclase-cAMP-PKA pathway and thereby activated Src by inhibiting its regulatory C-terminal Src kinase (Csk). We suggest a novel model of regulation of NMDARs by Gi/o-coupled receptors whereby inhibition of the cAMP-PKA pathway via mGluR2/3 activates Src kinase and potentiates GluN2A-containing NMDAR currents. This represents a potentially novel mechanism to correct the hypoglutamatergic state found in schizophrenia.


Assuntos
Receptores de Glutamato Metabotrópico/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Quinases da Família src/metabolismo , Adenilil Ciclases/metabolismo , Animais , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Técnicas In Vitro , N-Metilaspartato/farmacologia , Técnicas de Patch-Clamp , Ratos , Ratos Wistar , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Sinapses/fisiologia
11.
Adv Exp Med Biol ; 961: 433-47, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23224901

RESUMO

Stroke is a very strong risk factor for dementia. Furthermore, ischemic stroke and Alzheimer's disease (AD) share a number of overlapping mechanisms of neuron loss and dysfunction, including those induced by the inappropriate activation of N-methyl-D-aspartate receptors (NMDARs). These receptors form a major subtype of excitatory glutamate receptor. They are nonselective cation channels with appreciable Ca(2+) permeability, and their overactivation leads to neurotoxicity in the cortex and hippocampus. NMDARs have therefore been therapeutic targets in both conditions, but they have failed in the treatment of stroke, and there is limited rationale for using them in treating AD. In this chapter, we discuss current understanding of subtypes of NMDARs and their potential roles in -ischemic stroke and AD. We also discuss the properties of several other nonselective cation channels, transient receptor potential melastatin 2 and 7 channels, and their implications in linking these conditions.


Assuntos
Envelhecimento/metabolismo , Doença de Alzheimer/metabolismo , Isquemia Encefálica/metabolismo , Hipocampo/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Canais de Cátion TRPM/metabolismo , Envelhecimento/patologia , Doença de Alzheimer/patologia , Animais , Isquemia Encefálica/patologia , Cálcio/metabolismo , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Hipocampo/patologia , Humanos , Proteínas Serina-Treonina Quinases , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia
12.
Proc Natl Acad Sci U S A ; 109(43): 17651-6, 2012 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-23045697

RESUMO

Basal forebrain cholinergic neurons, which innervate the hippocampus and cortex, have been implicated in many forms of cognitive function. Immunolesion-based methods in animal models have been widely used to study the role of acetylcholine (ACh) neurotransmission in these processes, with variable results. Cholinergic neurons have been shown to release both glutamate and ACh, making it difficult to deduce the specific contribution of each neurotransmitter on cognition when neurons are eliminated. Understanding the precise roles of ACh in learning and memory is critical because drugs that preserve ACh are used as treatment for cognitive deficits. It is therefore important to define which cholinergic-dependent behaviors could be improved pharmacologically. Here we investigate the contributions of forebrain ACh on hippocampal synaptic plasticity and cognitive behavior by selective elimination of the vesicular ACh transporter, which interferes with synaptic storage and release of ACh. We show that elimination of vesicular ACh transporter in the hippocampus results in deficits in long-term potentiation and causes selective deficits in spatial memory. Moreover, decreased cholinergic tone in the forebrain is linked to hyperactivity, without changes in anxiety or depression-related behavior. These data uncover the specific contribution of forebrain cholinergic tone for synaptic plasticity and behavior. Moreover, these experiments define specific cognitive functions that could be targeted by cholinergic replacement therapy.


Assuntos
Comportamento Animal , Potenciação de Longa Duração , Memória , Prosencéfalo/metabolismo , Proteínas Vesiculares de Transporte de Acetilcolina/metabolismo , Animais , Western Blotting , Imunofluorescência , Aprendizagem , Camundongos , Camundongos Knockout , Plasticidade Neuronal , Reação em Cadeia da Polimerase
13.
Mol Brain ; 5: 11, 2012 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-22487454

RESUMO

BACKGROUND: Glutathione (GSH) plays an important role in neuronal oxidant defence. Depletion of cellular GSH is observed in neurodegenerative diseases and thereby contributes to the associated oxidative stress and Ca2+ dysregulation. Whether depletion of cellular GSH, associated with neuronal senescence, directly influences Ca2+ permeation pathways is not known. Transient receptor potential melastatin type 2 (TRPM2) is a Ca2+ permeable non-selective cation channel expressed in several cell types including hippocampal pyramidal neurons. Moreover, activation of TRPM2 during oxidative stress has been linked to cell death. Importantly, GSH has been reported to inhibit TRPM2 channels, suggesting they may directly contribute to Ca2+ dysregulation associated with neuronal senescence. Herein, we explore the relation between cellular GSH and TRPM2 channel activity in long-term cultures of hippocampal neurons. RESULTS: In whole-cell voltage-clamp recordings, we observe that TRPM2 current density increases in cultured pyramidal neurons over time in vitro. The observed increase in current density was prevented by treatment with NAC, a precursor to GSH synthesis. Conversely, treatment of cultures maintained for 2 weeks in vitro with L-BSO, which depletes GSH by inhibiting its synthesis, augments TRPM2 currents. Additionally, we demonstrate that GSH inhibits TRPM2 currents through a thiol-independent mechanism, and produces a 3.5-fold shift in the dose-response curve generated by ADPR, the intracellular agonist for TRPM2. CONCLUSION: These results indicate that GSH plays a physiologically relevant role in the regulation of TRPM2 currents in hippocampal pyramidal neurons. This interaction may play an important role in aging and neurological diseases associated with depletion of GSH.


Assuntos
Senescência Celular , Glutationa/metabolismo , Homeostase , Ativação do Canal Iônico , Neurônios/citologia , Células Piramidais/metabolismo , Canais de Cátion TRPM/metabolismo , Acetilcisteína/farmacologia , Adenosina Difosfato Ribose/farmacologia , Animais , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Glutationa/farmacologia , Células HEK293 , Homeostase/efeitos dos fármacos , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Células Piramidais/citologia , Células Piramidais/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Compostos de Sulfidrila/metabolismo , Canais de Cátion TRPM/genética , Fatores de Tempo
14.
FEBS J ; 279(1): 12-9, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21985328

RESUMO

The phosphorylation and trafficking of N-methyl-d-aspartate (NMDA) receptors are tightly regulated by the Src family tyrosine kinase Fyn, through dynamic interactions with various scaffolding proteins in the NMDA receptor complex. Fyn acts as a point of convergence for many signaling pathways that upregulate GluN2B-containing NMDA receptors. In the following review, we focus on Fyn signaling downstream of different G-protein-coupled receptors: the dopamine D1 receptor, and receptors cognate to the pituitary adenylate cyclase-activating polypeptide. The net result of activation of each of these signaling pathways is upregulation of GluN2B-containing NMDA receptors. The NMDA receptor is a major target of ethanol in the brain, and accumulating evidence suggests that Fyn mediates the effects of ethanol by regulating the phosphorylation of GluN2B NMDA receptor subunits. Furthermore, Fyn has been shown to regulate alcohol withdrawal and acute tolerance to ethanol through a GluN2B-dependent mechanism. In addition to its effects on NMDA receptor function, Fyn also modifies the threshold for synaptic plasticity at CA1 synapses, an effect that probably contributes to the effects of Fyn on spatial and contextual fear learning.


Assuntos
Regulação da Expressão Gênica , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Transdução de Sinais , Animais , Humanos , Plasticidade Neuronal , Receptores de N-Metil-D-Aspartato/metabolismo
15.
Neural Regen Res ; 7(10): 772-7, 2012 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-25737701

RESUMO

As the major division of the basal ganglia, neostriatum forms mutual connections with multiple brain areas and is critically involved in motor control and learning/memory. Long-term synaptic plasticity has been widely studied in different species recently. However, there are rare reports about the short-term synaptic plasticity in neostratium. In the present study, using field excitatory postsynaptic potentials recording, we reported one form of short-term synaptic plasticity that is paired pulse depression in juvenile rat dorsal striatum slices induced by stimuli of the white matter. The field excitatory postsynaptic potentials could be abolished by α-amino-3-hydroxy-5-methylizoxazole-4-propionic acid receptor antagonist, 6-cyano-7-nitroquinoxaline-2,3-dione, but not by gamma-aminobutyric acid type A receptor antagonist bicuculline or dopamine D1 receptor antagonist SKF-81297. The paired pulse depression in the corticostratial pathway was different from paired pulse facilitation in the hippocampal CA1 synapse. In addition, the paired pulse depression was not affected by bath application of gamma-aminobutyric acid type A receptor antagonist or dopamine D1 receptor antagonist. However, low calcium and high magnesium could attenuate the paired pulse depression. These findings suggest a more complicated plasticity form in the dorsal striatum of juvenile rats that is different from that in the hippocampus, which is related with extracellular calcium.

16.
EMBO J ; 31(4): 805-16, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22187052

RESUMO

Metaplasticity is a higher form of synaptic plasticity that is essential for learning and memory, but its molecular mechanisms remain poorly understood. Here, we report that metaplasticity of transmission at CA1 synapses in the hippocampus is mediated by Src family kinase regulation of NMDA receptors (NMDARs). We found that stimulation of G-protein-coupled receptors (GPCRs) regulated the absolute contribution of GluN2A-versus GluN2B-containing NMDARs in CA1 neurons: pituitary adenylate cyclase activating peptide 1 receptors (PAC1Rs) selectively recruited Src kinase, phosphorylated GluN2ARs, and enhanced their functional contribution; dopamine 1 receptors (D1Rs) selectively stimulated Fyn kinase, phosphorylated GluN2BRs, and enhanced these currents. Surprisingly, PAC1R lowered the threshold for long-term potentiation while long-term depression was enhanced by D1R. We conclude that metaplasticity is gated by the activity of GPCRs, which selectively target subtypes of NMDARs via Src kinases.


Assuntos
Receptores de N-Metil-D-Aspartato/metabolismo , Quinases da Família src/metabolismo , Animais , Western Blotting , Potenciais Pós-Sinápticos Excitadores , Hipocampo/metabolismo , Hipocampo/fisiologia , Imunoprecipitação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Plasticidade Neuronal , Fosforilação , Ratos , Ratos Wistar
17.
Artigo em Inglês | MEDLINE | ID: mdl-22162781

RESUMO

Futhan is a serine protease inhibitor and medicine in the treatment of disseminated intravascular coagulation (DIC) and acute pancreatitis. It is metabolized quickly in vivo. Here we show that Futhan reversibly inhibits NMDA receptors in hippocampal neurons and GABA(A) receptors both in hippocampal neurons and in A549 cells, a human alveolar epithelial cell line. The effect of Futhan on GABA(A) receptors in A549 cells is much more potent than its effect on GABA(A) receptors in hippocampal neurons (IC(50): 0.9 µM V.S. 7.3 µM). Since GABA(A) receptors are also expressed in various non-neuronal tissues, particularly in airway epithelia and GABA promotes mucus production during asthma, our findings indicate that Futhan may be developed as a novel aerosolized therapeutic to treat asthma through blocking GABA(A) receptors in the lung.

18.
Mol Brain ; 4: 44, 2011 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-22188973

RESUMO

Transient receptor potential melastatin 2 (TRPM2) is a calcium permeable non-selective cation channel that functions as a sensor of cellular redox status. Highly expressed within the CNS, we have previously demonstrated the functional expression of these channels in CA1 pyramidal neurons of the hippocampus. Although implicated in oxidative stress-induced neuronal cell death, and potentially in neurodegenerative disease, the physiological role of TRPM2 in the central nervous system is unknown. Interestingly, we have shown that the activation of these channels may be sensitized by co-incident NMDA receptor activation, suggesting a potential contribution of TRPM2 to synaptic transmission. Using hippocampal cultures and slices from TRPM2 null mice we demonstrate that the loss of these channels selectively impairs NMDAR-dependent long-term depression (LTD) while sparing long-term potentiation. Impaired LTD resulted from an inhibition of GSK-3ß, through increased phosphorylation, and a reduction in the expression of PSD95 and AMPARs. Notably, LTD could be rescued in TRPM2 null mice by recruitment of GSK-3ß signaling following dopamine D2 receptor stimulation. We propose that TRPM2 channels play a key role in hippocampal synaptic plasticity.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Hipocampo/citologia , N-Metilaspartato/metabolismo , Plasticidade Neuronal/fisiologia , Sinapses/metabolismo , Canais de Cátion TRPM/metabolismo , Animais , Células Cultivadas , Glicogênio Sintase Quinase 3 beta , Hipocampo/metabolismo , Camundongos , Camundongos Knockout , Neurônios/citologia , Neurônios/fisiologia , Técnicas de Patch-Clamp , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transmissão Sináptica/fisiologia , Canais de Cátion TRPM/genética
19.
J Alzheimers Dis ; 27(2): 243-52, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21799250

RESUMO

Alzheimer's disease (AD) is the most common form of dementia characterized by the presence of amyloid-ß (Aß) plaques and neurofibrillary tangles. The mechanisms leading to AD are not completely understood; however, recent evidence suggests that alterations in Fyn, a Src family kinase, might contribute to AD pathogenesis. A number of studies have demonstrated that Fyn is involved in synaptic plasticity, a cellular mechanism for learning and memory. In addition, Fyn plays a role in the regulation of Aß production and mediates Aß-induced synaptic deficits and neurotoxicity. Fyn also induces tyrosine phosphorylation of tau. Although many studies have implicated a role for Fyn in AD, the precise cellular and molecular mechanisms require further investigation. Novel insights into the role of Fyn in AD may help identify alternative pharmacological approaches for the treatment of AD.


Assuntos
Doença de Alzheimer/enzimologia , Sistemas de Liberação de Medicamentos , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Animais , Sistemas de Liberação de Medicamentos/tendências , Humanos , Emaranhados Neurofibrilares/enzimologia , Fosforilação/fisiologia , Placa Amiloide/tratamento farmacológico , Placa Amiloide/enzimologia , Proteínas Proto-Oncogênicas c-fyn/fisiologia
20.
PLoS One ; 6(7): e21970, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21789198

RESUMO

Together, acid-sensing ion channels (ASICs) and epithelial sodium channels (ENaC) constitute the majority of voltage-independent sodium channels in mammals. ENaC is regulated by a chloride channel, the cystic fibrosis transmembrane conductance regulator (CFTR). Here we show that ASICs were reversibly inhibited by activation of GABA(A) receptors in murine hippocampal neurons. This inhibition of ASICs required opening of the chloride channels but occurred with both outward and inward GABA(A) receptor-mediated currents. Moreover, activation of the GABA(A) receptors modified the pharmacological features and kinetic properties of the ASIC currents, including the time course of activation, desensitization and deactivation. Modification of ASICs by open GABA(A) receptors was also observed in both nucleated patches and outside-out patches excised from hippocampal neurons. Interestingly, ASICs and GABA(A) receptors interacted to regulate synaptic plasticity in CA1 hippocampal slices. The activation of glycine receptors, which are similar to GABA(A) receptors, also modified ASICs in spinal neurons. We conclude that GABA(A) receptors and glycine receptors modify ASICs in neurons through mechanisms that require the opening of chloride channels.


Assuntos
Canais de Cloreto/metabolismo , Canais Iônicos de Abertura Ativada por Ligante/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Canais de Sódio/metabolismo , Canais Iônicos Sensíveis a Ácido , Animais , Cloretos/metabolismo , Hipocampo/citologia , Técnicas In Vitro , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Íons , Cinética , Camundongos , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Técnicas de Patch-Clamp , Receptores de GABA-A/metabolismo , Receptores de Glicina/metabolismo , Ácido gama-Aminobutírico/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...