Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 3: 926, 2013.
Article in English | MEDLINE | ID: mdl-23378895

ABSTRACT

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.


Subject(s)
Receptors, Metabotropic Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , src-Family Kinases/metabolism , Adenylyl Cyclases/metabolism , Animals , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Hippocampus/drug effects , Hippocampus/physiology , In Vitro Techniques , N-Methylaspartate/pharmacology , Patch-Clamp Techniques , Rats , Rats, Wistar , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Synapses/physiology
2.
FEBS J ; 279(1): 12-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21985328

ABSTRACT

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.


Subject(s)
Gene Expression Regulation , Proto-Oncogene Proteins c-fyn/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Signal Transduction , Animals , Humans , Neuronal Plasticity , Receptors, N-Methyl-D-Aspartate/metabolism
3.
EMBO J ; 31(4): 805-16, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22187052

ABSTRACT

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.


Subject(s)
Receptors, N-Methyl-D-Aspartate/metabolism , src-Family Kinases/metabolism , Animals , Blotting, Western , Excitatory Postsynaptic Potentials , Hippocampus/metabolism , Hippocampus/physiology , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuronal Plasticity , Phosphorylation , Rats , Rats, Wistar
4.
J Alzheimers Dis ; 27(2): 243-52, 2011.
Article in English | MEDLINE | ID: mdl-21799250

ABSTRACT

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.


Subject(s)
Alzheimer Disease/enzymology , Drug Delivery Systems , Proto-Oncogene Proteins c-fyn/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Animals , Drug Delivery Systems/trends , Humans , Neurofibrillary Tangles/enzymology , Phosphorylation/physiology , Plaque, Amyloid/drug therapy , Plaque, Amyloid/enzymology , Proto-Oncogene Proteins c-fyn/physiology
5.
Hippocampus ; 21(10): 1053-61, 2011 Oct.
Article in English | MEDLINE | ID: mdl-20865743

ABSTRACT

The induction of long-term potentiation (LTP) of CA3-CA1 synapses requires activation of postsynaptic N-methyl-D-aspartate receptors (GluNRs). At resting potential, the contribution of GluNRs is limited by their voltage-dependent block by extracellular Mg(2+). High-frequency afferent stimulation is required to cause sufficient summation of excitatory synaptic potentials (EPSPs) to relieve this block and to permit an influx of Ca(2+). It has been assumed that this relief of Mg(2+) block is sufficient for induction. We postulated that the induction of LTP also requires a Src-dependent plasticity of GluNRs. Using whole-cell recordings, LTP (GluARs) of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors-EPSCS was induced by pairing postsynaptic depolarization with presynaptic stimulation. This LTP was both GluNR and Src-dependent, being sensitive to AP-5, a GluNR selective antagonist, or to SU6656, a Src-selective inhibitor. When CNQX was used to block all GluARs, we observed a long-lasting potentiation of GluNR-mediated EPSCs. This plasticity was prevented by transiently blocking GluNRs during the induction protocol or by chelating intracellular Ca(2+). GluNRs plasticity was also prevented by bath applications of SU6656 or intracellular applications of the Src-selective inhibitory peptide, Src(40-58). It was also blocked by preventing activation of protein kinase C, a kinase that is upstream of Src-kinase-dependent regulation of GluNRs. Both GluN2A and GluN2B receptors were found to contribute to the plasticity of GluNRs. The contribution of GluNRs and, in particular, their plasticity to the maintenance of LTP was explored using AP5 and SU6656, respectively. When applied >20 min after induction neither drug influenced the magnitude of LTP. However, when applied immediately after induction, treatment with either drug caused the initial magnitude of LTP to progressively decrease to a sustained phase of reduced amplitude. Collectively, our findings suggest that GluNR plasticity, although not strictly required for induction, is necessary for the maintenance of a nondecrementing component of LTP.


Subject(s)
Hippocampus/physiology , Long-Term Potentiation/physiology , Neuronal Plasticity/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/physiology , src-Family Kinases/metabolism , 2-Amino-5-phosphonovalerate/pharmacology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Excitatory Amino Acid Agonists/pharmacology , Excitatory Postsynaptic Potentials/physiology , Hippocampus/drug effects , Indoles/pharmacology , Long-Term Potentiation/drug effects , Male , Membrane Potentials/physiology , Neuronal Plasticity/drug effects , Patch-Clamp Techniques , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, Presynaptic/drug effects , Sulfonamides/pharmacology , Synapses/drug effects , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology , src-Family Kinases/antagonists & inhibitors
6.
J Alzheimers Dis ; 21(4): 1089-99, 2010.
Article in English | MEDLINE | ID: mdl-21504126

ABSTRACT

Alzheimer's disease (AD) is currently treated with cholinergic and glutamatergic therapies, which provide symptomatic benefit but do not reverse the underlying pathology or cognitive deficits. The prevalence of AD is expected to triple over the next 50 years, creating an urgency to develop effective "disease-modifying" therapies to reduce the economic burden of this devastating disorder. One of the main areas of therapeutic focus has been an antiinflammatory strategy based on an inflammatory hypothesis of AD. This hypothesis originated from epidemiological evidence that long-term exposure to nonsteroidal anti-inflammatory drugs (NSAIDs) protected against the development of AD. However, large-scale double-blind placebo-controlled clinical trials have not supported the use of NSAIDS in treating AD. The following review outlines epidemiological, preclinical, and clinical evidence evaluating the efficacy of various NSAIDs and selective COX-2 inhibitors in AD. We also review recent anecdotal data with the TNF-α inhibitor, etanercept, and discuss possible explanations for the failure of preclinical data to translate into successful clinical trials.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cyclooxygenase 2 Inhibitors/therapeutic use , Encephalitis/drug therapy , Encephalitis/pathology , Neurons/drug effects , Neurons/pathology , Animals , Clinical Trials as Topic/methods , Drug Evaluation, Preclinical/methods , Humans
7.
Pediatr Res ; 65(5): 499-503, 2009 May.
Article in English | MEDLINE | ID: mdl-19190545

ABSTRACT

Infantile spasms is a catastrophic childhood seizure disorder for which few animal models exist. Children with Down syndrome are highly susceptible to infantile spasms. The Ts65Dn mouse is a valid model for Down syndrome; therefore, we tested the hypothesis that the Ts65Dn mouse represents a substrate for an animal model of infantile spasms. The baseline of naïve Ts65Dn mice showed spontaneous spike-and-wave discharges, a pattern that worsened with baclofen and gamma-butyrolactone, which induced acute epileptic extensor spasms (AEES) associated with epileptiform polyspike bursts and an electrodecremental response on the EEG. GABABR-agonist-induced AEES were significantly reduced with vigabatrin, rodent ACTH fragment, valproic acid, ethosuximide, and CGP 35348. Porcine ACTH had no effect. GABABR protein expression was significantly increased in the thalamus and medulla oblongata of Ts65D mice in comparison with wild-type controls. The GABABR agonist-treated Ts65Dn mouse shows the unique clinical, electrographic, and pharmacologic signature of infantile spasms and represents a valid, acute model of this disorder. GABABR-mediated mechanisms may contribute to the increased susceptibility of children with Down syndrome to infantile spasms.


Subject(s)
4-Butyrolactone , Baclofen , Brain/metabolism , Down Syndrome/complications , GABA Agonists , Receptors, GABA-B/metabolism , Spasms, Infantile/chemically induced , Animals , Anticonvulsants/pharmacology , Brain/drug effects , Brain/physiopathology , Disease Models, Animal , Down Syndrome/drug therapy , Down Syndrome/genetics , Down Syndrome/physiopathology , Electroencephalography , Ethosuximide/pharmacology , Genotype , Humans , Infant , Mice , Mice, Mutant Strains , Organophosphorus Compounds/pharmacology , Phenotype , Receptors, GABA-A/drug effects , Receptors, GABA-A/metabolism , Receptors, GABA-B/drug effects , Spasms, Infantile/genetics , Spasms, Infantile/physiopathology , Spasms, Infantile/prevention & control , Time Factors , Valproic Acid/pharmacology , Vigabatrin/pharmacology
8.
Hippocampus ; 19(9): 779-89, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19173226

ABSTRACT

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.


Subject(s)
Hippocampus/physiology , Neurons/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission/physiology , Vasoactive Intestinal Peptide/metabolism , Adenylyl Cyclases/metabolism , Animals , Cell Membrane/physiology , Cells, Cultured , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , In Vitro Techniques , Male , Membrane Potentials/physiology , Patch-Clamp Techniques , Rats , Rats, Wistar , Receptors, Vasoactive Intestinal Peptide/antagonists & inhibitors , Signal Transduction , Thionucleotides/pharmacology , Time Factors
9.
J Biol Chem ; 284(12): 8054-63, 2009 Mar 20.
Article in English | MEDLINE | ID: mdl-19106110

ABSTRACT

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.


Subject(s)
Hippocampus/metabolism , N-Methylaspartate/metabolism , Neurons/metabolism , Platelet-Derived Growth Factor/pharmacology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Actins/metabolism , Animals , Becaplermin , Calcium/metabolism , Cyclic AMP Response Element-Binding Protein , Cytoskeleton/metabolism , Hippocampus/cytology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/cytology , Phenols/pharmacology , Phospholipase C gamma/metabolism , Piperidines/pharmacology , Platelet-Derived Growth Factor/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-sis , Rats , Rats, Wistar , Receptor, Platelet-Derived Growth Factor beta/agonists , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...