Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
Eur J Pharmacol ; 648(1-3): 15-23, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20854810

RESUMO

Acid sensing ion channels (ASICs) are implicated in various brain functions including learning and memory and are involved in a number of neurological disorders such as pain, ischemic stroke, depression, and multiple sclerosis. We have recently defined ASICs as one of receptor targets of aromatic diamidines in neurons. Aromatic diamidines are DNA-binding agents and have long been used in the treatment of leishmaniasis, trypanosomiasis, pneumocystis pneumonia and babesiosis. Moreover, some aromatic diamidines are used as skin-care and baby products and others have potential to suppress tumor growth or to combat malaria. A large number of aromatic diamidines or analogs have been synthesized. Many efforts are being made to optimize the therapeutic spectrum of aromatic diamidines, i.e. to reduce toxicity, increase oral bioavailability and enhance their penetration of the blood-brain barrier. Aromatic diamidines therefore provide a shortcut of screening for selective ASIC inhibitors with therapeutic potential. Intriguingly nafamostat, a protease inhibitor for treating acute pancreatitis, also inhibits ASIC activities. Aromatic diamidines and nafamostat have many similarities although they belong to distinct classes of medicinal agents for curing different diseases. Here we delineate background, clinical application and drug development of aromatic diamidines that could facilitate the screening for selective ASIC inhibitors for research purposes. Further studies may lead to a drug with therapeutic value and extend the therapeutic scope of aromatic diamidines to combat neurological diseases.


Assuntos
Amidinas/química , Amidinas/farmacologia , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Proteínas do Tecido Nervoso/antagonistas & inibidores , Doenças do Sistema Nervoso/tratamento farmacológico , Canais Iônicos Sensíveis a Ácido , Amidinas/síntese química , Amidinas/uso terapêutico , Sequência de Aminoácidos , Animais , Humanos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Doenças do Sistema Nervoso/metabolismo , Canais de Sódio/química , Canais de Sódio/metabolismo
10.
J Mol Neurosci ; 42(3): 319-26, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20414742

RESUMO

Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP) are two closely related peptides, which can activate protein kinase A (PKA). At least three receptors for PACAP and VIP have been identified. The PACAP-specific receptor, PAC1 receptor, exhibits a higher affinity for PACAP than VIP, whereas VIP receptors, VPAC1-R and VPAC2-R, have similar affinities for PACAP and VIP. Both PACAP/VIP and their cognate receptors are highly expressed in the brain, including the hippocampus. Recently, their roles in the regulation of synaptic transmission have begun to emerge. PACAP/VIP can signal through different pathways to regulate N-methyl-D: -aspartate (NMDA) receptors in CA1 pyramidal cells. The activation of VPAC1/2-Rs increases evoked NMDA currents via the cyclic AMP/PKA pathway. However, the activation of PAC1-R stimulates a PLC/PKC/Pyk2/Src signaling pathway to enhance NMDA receptor function in hippocampal neurons. Furthermore, different concentrations of PACAP induce different effects on the both α-amino-3-hydroxy-5-isoxazole-propionic acid-evoked current and basal synaptic transmission by activating different receptors. Their roles in learning and memory are also demonstrated using transgenic mice and pharmacological methods.


Assuntos
Hipocampo/fisiologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Transmissão Sináptica/fisiologia , Peptídeo Intestinal Vasoativo/metabolismo , Animais , Hipocampo/citologia , Aprendizagem/fisiologia , Memória/fisiologia , Camundongos , N-Metilaspartato/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Receptores de Peptídeo Intestinal Vasoativo/metabolismo , Transdução de Sinais/fisiologia , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/metabolismo
11.
Int J Physiol Pathophysiol Pharmacol ; 2(2): 95-103, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21383889

RESUMO

NMDA receptor overactivation triggers intracellular Ca(2+) dysregulation, which has long been thought to be critical for initiating excitotoxic cell death cascades associated with stroke and neurodegenerative disease. The inability of NMDA receptor antagonists to afford neuroprotection in clinical stroke trials has led to a re-evaluation of excitotoxic models of cell death and has focused research efforts towards identifying additional Ca(2+) influx pathways. Recent studies indicate that TRPM2, a member of the TRPM subfamily of Ca(2+)-permeant, non-selective cation channel, plays an important role in mediating cellular responses to a wide range of stimuli that, under certain situations, can induce cell death. These include reactive oxygen and nitrogen species, tumour necrosis factor as well as soluble oli-gomers of amyloid beta. However, the molecular basis of TRPM2 channel involvement in these processes is not fully understood. In this review, we summarize recent studies about the regulation of TRPM2, its interaction with calcium and the possible implications for neurodegenerative diseases.

12.
Stem Cells Dev ; 18(10): 1423-32, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19355840

RESUMO

Transplantation of neural stem/progenitor cells (NSPCs) is a promising strategy for repair of the diseased/injured central nervous system (CNS); however, controlling their differentiation remains a significant hurdle. This study is aimed at controlling differentiation and specifically at screening exogenous factors to direct NSPC differentiation into neurons in vitro. In this study, adult rat SVZ-derived NSPCs were treated with several factors and screened individually and in combination for changes in cellular morphology, neuronal marker expression, quantitative real-time qRT-PCR, and electrophysiological properties. These in vitro screens showed that of all the different treatments, dibutyryl cyclic AMP (dbcAMP) and interferon-gamma (IFN-gamma) enhanced neuronal differentiation most significantly compared to the 1% fetal bovine serum (FBS) controls. Importantly, the combined treatment of NSPCs with dbcAMP and IFN-gamma promoted greater neuronal differentiation as reflected by an increase in beta-III tubulin expression and morphological differentiation. Interestingly, the neurons that were generated from the NSPCs in vitro in the presence of dbcAMP and IFN-gamma, alone or in combination, responded to exogenous glutamate (Glu), but not gamma-aminobutyric acid (GABA), indicating that these neurons express glutamate receptors. These NSPC-derived neurons may be promising for neural regenerative strategies in the CNS.


Assuntos
Bucladesina/farmacologia , Diferenciação Celular/efeitos dos fármacos , Interferon gama/farmacologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Envelhecimento/efeitos dos fármacos , Animais , Sobrevivência Celular/efeitos dos fármacos , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Neurônios/metabolismo , Prosencéfalo/citologia , Ratos , Ratos Wistar , Células-Tronco/metabolismo
13.
J Physiol ; 587(Pt 5): 965-79, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19124544

RESUMO

TRPM2 is a Ca(2+)-permeable member of the transient receptor potential melastatin family of cation channels whose activation by reactive oxygen/nitrogen species (ROS/RNS) and ADP-ribose (ADPR) is linked to cell death. While these channels are broadly expressed in the CNS, the presence of TRPM2 in neurons remains controversial and more specifically, whether they are expressed in neurons of the hippocampus is an open question. With this in mind, we examined whether functional TRPM2 channels are expressed in this neuronal population. Using a combination of molecular and biochemical approaches, we demonstrated the expression of TRPM2 transcripts and proteins in hippocampal pyramidal neurons. Whole-cell voltage-clamp recordings were subsequently carried out to assess the presence of TRPM2-mediated currents. Application of hydrogen peroxide or peroxynitrite to cultured hippocampal pyramidal neurons activated an inward current that was abolished upon removal of extracellular Ca(2+), a hallmark of TRPM2 activation. When ADPR (300 microM) was included in the patch pipette, a large inward current developed but only when depolarizing voltage ramps were continuously (1/10 s) applied to the membrane. This current exhibited a linear current-voltage relationship and was sensitive to block by TRPM2 antagonists (i.e. clotrimazole, flufenamic acid and N-(p-amylcinnamoyl)anthranilic acid (ACA)). The inductive effect of voltage ramps on the ADPR-dependent current required voltage-dependent Ca(2+) channels (VDCCs) and a rise in [Ca(2+)](i). Consistent with the need for a rise in [Ca(2+)](i), activation of NMDA receptors (NMDARs), which are highly permeable to Ca(2+), was also permissive for current development. Importantly, given the prominent vulnerability of CA1 neurons to free-radical-induced cell death, we confirmed that, with ADPR in the pipette, a brief application of NMDA could evoke a large inward current in CA1 pyramidal neurons from hippocampal slices that was abolished by the removal of extracellular Ca(2+), consistent with TRPM2 activation. Such a current was absent in interneurons of CA1 stratum radiatum. Finally, infection of cultured hippocampal neurons with a TRPM2-specific short hairpin RNA (shRNA(TRPM2)) significantly reduced both the expression of TRPM2 and the amplitude of the ADPR-dependent current. Taken together, these results indicate that hippocampal pyramidal neurons possess functional TRPM2 channels whose activation by ADPR is functionally coupled to VDCCs and NMDARs through a rise in [Ca(2+)](i).


Assuntos
Cálcio/metabolismo , Clusterina/biossíntese , Neurônios/metabolismo , Células Piramidais/metabolismo , Animais , Cálcio/antagonistas & inibidores , Cálcio/fisiologia , Bloqueadores dos Canais de Cálcio/farmacologia , Células Cultivadas , Clusterina/metabolismo , Clusterina/fisiologia , Feminino , Masculino , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Gravidez , Células Piramidais/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/fisiologia
14.
Hippocampus ; 19(9): 779-89, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19173226

RESUMO

Vasoactive intestinal peptide (VIP) is a 28-amino acid peptide, which belongs to a superfamily of structurally related peptide hormones including pituitary adenylate cyclase-activating polypeptide (PACAP). Although several studies have identified the involvement of PACAP in learning and memory, little work has been done to investigate such a role for VIP. At least three receptors for VIP have been identified including the PACAP receptor (PAC1-R) and the two VIP receptors (VPAC receptors). VIP can activate the PAC1-R only if it is used at relatively high concentrations (e.g., 100 nM); however, at lower concentrations (e.g., 1 nM) it is selective for the VPAC receptors. Our lab has showed that PAC1-R activation signals through PKC/CAKbeta/Src pathway to regulate NMDA receptors; however, there is little known about the potential regulation of NMDA receptors by VPAC receptors. Our studies demonstrated that application of 1 nM VIP enhanced NMDA currents by stimulating the VPAC receptors as the effect was blocked by VPAC receptor antagonist [Ac-Tyr(1), D-Phe(2)]GRF (1-29). This enhancement of NMDA currents was blocked by both Rp-cAMPS and PKI(14-22) (they are highly specific PKA inhibitors), but not by the specific PKC inhibitor, bisindolylmaleimide I. In addition, the VIP-induced enhancement of NMDA currents was accentuated by inhibition of phosphodiesterase 4, which inhibits the degradation of cAMP. This regulation of NMDA receptors also required the scaffolding protein AKAP. In contrast, the potentiation induced by high concentration of VIP (e.g., 100 nM) was mediated by PAC1-R as well as by Src kinase. Overall, these results show that VIP can regulate NMDA receptors through different receptors and signaling pathways.


Assuntos
Hipocampo/fisiologia , Neurônios/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Transmissão Sináptica/fisiologia , Peptídeo Intestinal Vasoativo/metabolismo , Adenilil Ciclases/metabolismo , Animais , Membrana Celular/fisiologia , Células Cultivadas , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacologia , Técnicas In Vitro , Masculino , Potenciais da Membrana/fisiologia , Técnicas de Patch-Clamp , Ratos , Ratos Wistar , Receptores de Peptídeo Intestinal Vasoativo/antagonistas & inibidores , Transdução de Sinais , Tionucleotídeos/farmacologia , Fatores de Tempo
15.
J Biol Chem ; 284(12): 8054-63, 2009 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-19106110

RESUMO

Platelet-derived growth factor (PDGF) beta receptor activation inhibits N-methyl-d-aspartate (NMDA)-evoked currents in hippocampal and cortical neurons via the activation of phospholipase Cgamma, PKC, the release of intracellular calcium, and a rearrangement of the actin cytoskeleton. In the hippocampus, the majority of NMDA receptors are heteromeric; most are composed of 2 NR1 subunits and 2 NR2A or 2 NR2B subunits. Using NR2B- and NR2A-specific antagonists, we demonstrate that PDGF-BB treatment preferentially inhibits NR2B-containing NMDA receptor currents in CA1 hippocampal neurons and enhances long-term depression in an NR2B subunit-dependent manner. Furthermore, treatment of hippocampal slices or cultures with PDGF-BB decreases the surface localization of NR2B but not of NR2A subunits. PDGFbeta receptors colocalize to a higher degree with NR2B subunits than with NR2A subunits. After neuronal injury, PDGFbeta receptors and PDGF-BB are up-regulated and PDGFbeta receptor activation is neuroprotective against glutamate-induced neuronal damage in cultured neurons. We demonstrate that the neuroprotective effects of PDGF-BB are occluded by the NR2B antagonist, Ro25-6981, and that PDGF-BB promotes NMDA signaling to CREB and ERK1/2. We conclude that PDGFbetaR signaling, by preferentially targeting NR2B receptors, provides an important mechanism for neuroprotection by growth factors in the central nervous system.


Assuntos
Hipocampo/metabolismo , N-Metilaspartato/metabolismo , Neurônios/metabolismo , Fator de Crescimento Derivado de Plaquetas/farmacologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Actinas/metabolismo , Animais , Becaplermina , Cálcio/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Citoesqueleto/metabolismo , Hipocampo/citologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neurônios/citologia , Fenóis/farmacologia , Fosfolipase C gama/metabolismo , Piperidinas/farmacologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-sis , Ratos , Ratos Wistar , Receptor beta de Fator de Crescimento Derivado de Plaquetas/agonistas , Transdução de Sinais/efeitos dos fármacos
16.
Science ; 322(5907): 1555-9, 2008 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-19056988

RESUMO

Pannexin-1 (Px1) is expressed at postsynaptic sites in pyramidal neurons, suggesting that these hemichannels contribute to dendritic signals associated with synaptic function. We found that, in pyramidal neurons, N-methyl-d-aspartate receptor (NMDAR) activation induced a secondary prolonged current and dye flux that were blocked with a specific inhibitory peptide against Px1 hemichannels; knockdown of Px1 by RNA interference blocked the current in cultured neurons. Enhancing endogenous NMDAR activation in brain slices by removing external magnesium ions (Mg2+) triggered epileptiform activity, which had decreased spike amplitude and prolonged interburst interval during application of the Px1 hemichannel blocking peptide. We conclude that Px1 hemichannel opening is triggered by NMDAR stimulation and can contribute to epileptiform seizure activity.


Assuntos
Conexinas/fisiologia , Hipocampo/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Células Piramidais/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Potenciais de Ação , Trifosfato de Adenosina/metabolismo , Animais , Cálcio/metabolismo , Células Cultivadas , Conexinas/genética , Dendritos/fisiologia , Sinapses Elétricas/fisiologia , Epilepsia/fisiopatologia , Hipocampo/fisiopatologia , Técnicas In Vitro , Camundongos , Proteínas do Tecido Nervoso/genética , Técnicas de Patch-Clamp , Interferência de RNA , Ratos , Ratos Wistar , Transmissão Sináptica
17.
Mol Brain ; 1: 20, 2008 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-19077273

RESUMO

BACKGROUND: We have previously demonstrated that PDGF receptor activation indirectly inhibits N-methyl-D-aspartate (NMDA) currents by modifying the cytoskeleton. PDGF receptor ligand is also neuroprotective in hippocampal slices and cultured neurons. PDGF receptors are tyrosine kinases that control a variety of signal transduction pathways including those mediated by PLCγ. In fibroblasts Src and another non-receptor tyrosine kinase, Abelson kinase (Abl), control PDGF receptor regulation of cytoskeletal dynamics. The mechanism whereby PDGF receptor regulates cytoskeletal dynamics in central neurons remains poorly understood. RESULTS: Intracellular applications of active Abl, but not heat-inactivated Abl, decreased NMDA-evoked currents in isolated hippocampal neurons. This mimics the effects of PDGF receptor activation in these neurons. The Abl kinase inhibitor, STI571, blocked the inhibition of NMDA currents by Abl. We demonstrate that PDGF receptors can activate Abl kinase in hippocampal neurons via mechanisms similar to those observed previously in fibroblasts. Furthermore, PDGFß receptor activation alters the subcellular localization of Abl. Abl kinase is linked to actin cytoskeletal dynamics in many systems. We show that the inhibition of NMDA receptor currents by Abl kinase is blocked by the inclusion of the Rho kinase inhibitor, Y-27632, and that activation of Abl correlates with an increase in ROCK tyrosine phosphorylation. CONCLUSION: This study demonstrates that PDGFß receptors act via an interaction with Abl kinase and Rho kinase to regulated cytoskeletal regulation of NMDA receptor channels in CA1 pyramidal neurons.


Assuntos
Região CA1 Hipocampal/metabolismo , Citoesqueleto/metabolismo , Neurônios/enzimologia , Proteínas Proto-Oncogênicas c-abl/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Amidas/farmacologia , Animais , Becaplermina , Região CA1 Hipocampal/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Camundongos , Modelos Biológicos , Neurônios/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Proto-Oncogênicas c-abl/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-sis , Piridinas/farmacologia , Ratos , Ratos Wistar , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/metabolismo
18.
Biochim Biophys Acta ; 1768(4): 941-51, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17261268

RESUMO

Long-term potentiation (LTP) and long-term depression (LTD) are the major forms of functional synaptic plasticity observed at CA1 synapses of the hippocampus. The balance between LTP and LTD or "metaplasticity" is controlled by G-protein coupled receptors (GPCRs) whose signal pathways target the N-methyl-D-asparate (NMDA) subtype of excitatory glutamate receptor. We discuss the protein kinase signal cascades stimulated by Galphaq and Galphas coupled GPCRs and describe how control of NMDAR activity shifts the threshold for the induction of LTP.


Assuntos
Hipocampo/fisiologia , Plasticidade Neuronal , Receptores Acoplados a Proteínas G/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/fisiologia , Animais , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Hipocampo/citologia , Hipocampo/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Plasticidade Neuronal/genética , Neurônios/metabolismo , Neurônios/fisiologia , Proteínas Tirosina Quinases/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores de N-Metil-D-Aspartato/genética , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA