Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
NPJ Parkinsons Dis ; 10(1): 49, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38429321

ABSTRACT

Pathogenic mutations in LRRK2 cause Parkinson's disease (PD). The G2019S variant is the most common, which results in abnormally high kinase activity. Compounds that target LRRK2 kinase activity are currently being developed and tested in clinical trials. We recently found that G2019S LRRK2 causes mitochondrial DNA (mtDNA) damage and treatment with multiple classes of LRRK2 kinase inhibitors at concentrations associated with dephosphorylation of LRRK2 reversed mtDNA damage to healthy control levels. Because maintaining the normal function of LRRK2 in heterozygous G2019S LRRK2 carriers while specifically targeting the G2019S LRRK2 activity could have an advantageous safety profile, we explored the efficacy of a G2019S mutant selective LRRK2 inhibitor to reverse mtDNA damage in G2019S LRRK2 models and patient cells relative to non-selective LRRK2 inhibitors. Potency of LRRK2 kinase inhibition by EB-42168, a G2019S mutant LRRK2 kinase inhibitor, and MLi-2, a non-selective inhibitor, was determined by measuring phosphorylation of LRRK2 at Ser935 and/or Ser1292 using quantitative western immunoblot analysis. The Mito DNADX assay, which allows for the accurate real-time quantification of mtDNA damage in a 96-well platform, was performed in parallel. We confirmed that EB-42168 selectively inhibits LRRK2 phosphorylation on G2019S LRRK2 relative to wild-type LRRK2. On the other hand, MLi-2 was equipotent for wild-type and G2019S LRRK2. Acute treatment with EB-42168 inhibited LRRK2 phosphorylation and also restored mtDNA damage to healthy control levels. We further investigated the relationship between LRRK2 kinase activity, mtDNA damage and mitophagy. Levels of mtDNA damage caused by G2019S LRRK2 were fully re-established within 2 h of a LRRK2 inhibitor wash out and recovery experiment, indicating the mtDNA damage phenotype is highly dynamic. G2019S LRRK2 mitophagy defects were not alleviated with LRRK2 kinase inhibition, suggesting that mitophagy is not mechanistically regulating LRRK2 kinase-mediated reversal of mtDNA damage in this acute timeframe. Abrogation of mtDNA damage with the mutant selective tool inhibitor EB-42168 demonstrates the potential of a precision medicine approach for LRRK2 G2019S PD. Levels of mtDNA damage may serve as a potential pharmacodynamic biomarker of altered kinase activity that could be useful for small molecule development and clinical trials.

2.
Bioorg Med Chem Lett ; 95: 129487, 2023 Oct 15.
Article in English | MEDLINE | ID: mdl-37734423

ABSTRACT

The G2019S variant of LRRK2, which causes an increase in kinase activity, is associated with the occurrence of Parkinson's disease (PD). Potent, mutation-selective, and brain penetrant inhibitors of LRRK2 can suppress the biological effects specific to G2019S-LRRK2 that cause pathogenicity. We report the discovery of a series of cyanoindane and cyanotetralin kinase inhibitors culminating in compound 34 that demonstrated selective inhibition of phosphorylation of LRRK2 in the mouse brain. These novel inhibitors may further enable the precision medicine path for future PD therapeutics.

3.
Mov Disord ; 36(6): 1362-1371, 2021 06.
Article in English | MEDLINE | ID: mdl-33836114

ABSTRACT

BACKGROUND: A common genetic mutation that causes Parkinson's disease (PD) is the G2019S LRRK2 mutation. A precision medicine approach that selectively blocks only excess kinase activity of the mutant allele could yield a safe and effective treatment for G2019S LRRK2 PD. OBJECTIVE: To determine the activity of a G2019S mutant selective leucine-rich repeat kinase 2 (LRRK2) kinase inhibitor as compared to a nonselective inhibitor in blood of subjects with genetic and idiopathic PD on two LRRK2 biomarkers, pSer935 LRRK2 and pThr73 Rab10. METHODS: Blood was collected from 13 subjects with or without a G2019S LRRK2 mutation with PD and one healthy control. Peripheral blood mononuclear cells were treated ex vivo with a novel G2019S LRRK2 inhibitor (EB-42168) or the nonselective inhibitor MLi-2. Quantitative western immunoblot analyses were performed. RESULTS: EB-42168 was 100 times more selective for G2019S LRRK2 when compared to wild-type (WT) LRRK2. Concentrations that inhibited phosphorylation of pSer935 LRRK2 by 90% in homozygous G2019S LRRK2 patients, inhibited pSer935 LRRK2 by 36% in heterozygous patients, and by only 5% in patients carrying only the WT allele. Similar selectivity was seen for pThr73 Rab10. MLi-2 showed an equivalent level of inhibition across all genotypes. CONCLUSIONS: These findings demonstrate that EB-42168, a G2019S LRRK2 selective inhibitor, lowers mutant G2019S LRRK2 phosphorylated biomarkers while simultaneously sparing WT LRRK2. Selective targeting of G2019S LRRK2 with a small molecule lays the foundation for a precision medicine treatment of G2019S LRRK2 PD. © 2021 ESCAPE Bio, Inc. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Subject(s)
Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/antagonists & inhibitors , Parkinson Disease , Heterozygote , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leukocytes, Mononuclear , Mutation/genetics , Parkinson Disease/drug therapy , Parkinson Disease/genetics
4.
J Med Chem ; 63(23): 14821-14839, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33197196

ABSTRACT

Pathogenic variants in the leucine-rich repeat kinase 2 (LRRK2) gene have been identified that increase the risk for developing Parkinson's disease in a dominantly inherited fashion. These pathogenic variants, of which G2019S is the most common, cause abnormally high kinase activity, and compounds that inhibit this activity are being pursued as potentially disease-modifying therapeutics. Because LRRK2 regulates important cellular processes, developing inhibitors that can selectively target the pathogenic variant while sparing normal LRRK2 activity could offer potential advantages in heterozygous carriers. We conducted a high-throughput screen and identified a single selective compound that preferentially inhibited G2019S-LRRK2. Optimization of this scaffold led to a series of novel, potent, and highly selective G2019S-LRRK2 inhibitors.


Subject(s)
Indazoles/pharmacology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Tetrazoles/pharmacology , Animals , HEK293 Cells , High-Throughput Screening Assays , Humans , Indazoles/chemical synthesis , Indazoles/pharmacokinetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Mice , Molecular Structure , Mutation , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/chemical synthesis , Pyrimidines/pharmacokinetics , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/pharmacokinetics , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Tetrazoles/chemical synthesis , Tetrazoles/pharmacokinetics
5.
F1000Res ; 5: 137, 2016.
Article in English | MEDLINE | ID: mdl-26998235

ABSTRACT

A common pathological hallmark of age-related neurodegenerative diseases is the intracellular accumulation of protein aggregates such as α-synuclein in Parkinson's disease, TDP-43 in ALS, and tau in Alzheimer's disease. Enhancing intracellular clearance of aggregation-prone proteins is a plausible strategy for slowing progression of neurodegenerative diseases and there is great interest in identifying molecular targets that control protein turnover. One of the main routes for protein degradation is through the proteasome, a multisubunit protease that degrades proteins that have been tagged with a polyubiquitin chain by ubiquitin activating and conjugating enzymes. Published data from cellular models indicate that Ubiquitin-specific protease 14 (USP14), a deubiquitinating enzyme (DUB), slows the degradation of tau and TDP-43 by the proteasome and that an inhibitor of USP14 increases the degradation of these substrates. We conducted similar experiments designed to evaluate tau, TDP-43, or α-synuclein levels in cells after overexpressing USP14 or knocking down endogenous expression by siRNA.

6.
Autophagy ; 12(4): 713-26, 2016.
Article in English | MEDLINE | ID: mdl-27050463

ABSTRACT

Autophagy is the primary process for recycling cellular constituents through lysosomal degradation. In addition to nonselective autophagic engulfment of cytoplasm, autophagosomes can recognize specific cargo by interacting with ubiquitin-binding autophagy receptors such as SQSTM1/p62 (sequestosome 1). This selective form of autophagy is important for degrading aggregation-prone proteins prominent in many neurodegenerative diseases. We carried out a high content image-based siRNA screen (4 to 8 siRNA per gene) for modulators of autophagic flux by monitoring fluorescence of GFP-SQSTM1 as well as colocalization of GFP-SQSTM1 with LAMP2 (lysosomal-associated membrane protein 2)-positive lysosomal vesicles. GFP-SQSTM1 and LAMP2 phenotypes of primary screen hits were confirmed in 2 cell types and profiled with image-based viability and MTOR signaling assays. Common seed analysis guided siRNA selection for these assays to reduce bias toward off-target effects. Confirmed hits were further validated in a live-cell assay to monitor fusion of autophagosomes with lysosomes. Knockdown of 10 targets resulted in phenotypic profiles across multiple assays that were consistent with upregulation of autophagic flux. These hits include modulators of transcription, lysine acetylation, and ubiquitination. Two targets, KAT8 (K[lysine] acetyltransferase 8) and CSNK1A1 (casein kinase 1, α 1), have been implicated in autophagic regulatory feedback loops. We confirmed that CSNK1A1 knockout (KO) cell lines have accelerated turnover of long-lived proteins labeled with (14)C-leucine in a pulse-chase assay as additional validation of our screening assays. Data from this comprehensive autophagy screen point toward novel regulatory pathways that might yield new therapeutic targets for neurodegeneration.


Subject(s)
Autophagy , High-Throughput Screening Assays/methods , Imaging, Three-Dimensional , RNA, Small Interfering/metabolism , Animals , Cell Line, Tumor , Cell Survival , Gene Knockdown Techniques , Green Fluorescent Proteins/metabolism , Humans , Reproducibility of Results
7.
J Biol Chem ; 290(8): 4908-4927, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25533468

ABSTRACT

synGAP is a neuron-specific Ras and Rap GTPase-activating protein (GAP) found in high concentrations in the postsynaptic density (PSD) fraction from the mammalian forebrain. We have previously shown that, in situ in the PSD fraction or in recombinant form in Sf9 cell membranes, synGAP is phosphorylated by Ca(2+)/calmodulin-dependent protein kinase II (CaMKII), another prominent component of the PSD. Here, we show that recombinant synGAP (r-synGAP), lacking 102 residues at the N terminus, can be purified in soluble form and is phosphorylated by cyclin-dependent kinase 5 (CDK5) as well as by CaMKII. Phosphorylation of r-synGAP by CaMKII increases its HRas GAP activity by 25% and its Rap1 GAP activity by 76%. Conversely, phosphorylation by CDK5 increases r-synGAP's HRas GAP activity by 98% and its Rap1 GAP activity by 20%. Thus, phosphorylation by both kinases increases synGAP activity; CaMKII shifts the relative GAP activity toward inactivation of Rap1, and CDK5 shifts the relative activity toward inactivation of HRas. GAP activity toward Rap2 is not altered by phosphorylation by either kinase. CDK5 phosphorylates synGAP primarily at two sites, Ser-773 and Ser-802. Phosphorylation at Ser-773 inhibits r-synGAP activity, and phosphorylation at Ser-802 increases it. However, the net effect of concurrent phosphorylation of both sites, Ser-773 and Ser-802, is an increase in GAP activity. synGAP is phosphorylated at Ser-773 and Ser-802 in the PSD fraction, and its phosphorylation by CDK5 and CaMKII is differentially regulated by activation of NMDA-type glutamate receptors in cultured neurons.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Cyclin-Dependent Kinase 5 , GTPase-Activating Proteins , Oncogene Proteins , Proto-Oncogene Proteins p21(ras) , Synapses/enzymology , rap GTP-Binding Proteins , rap1 GTP-Binding Proteins , ras GTPase-Activating Proteins , ras Proteins , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/chemistry , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cells, Cultured , Cyclin-Dependent Kinase 5/chemistry , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Humans , Neurons/cytology , Neurons/enzymology , Oncogene Proteins/chemistry , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Phosphorylation , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Rats , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , rap1 GTP-Binding Proteins/chemistry , rap1 GTP-Binding Proteins/genetics , rap1 GTP-Binding Proteins/metabolism , ras GTPase-Activating Proteins/chemistry , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism , ras Proteins/chemistry , ras Proteins/genetics , ras Proteins/metabolism
8.
J Neurosci ; 31(45): 16194-207, 2011 Nov 09.
Article in English | MEDLINE | ID: mdl-22072671

ABSTRACT

Densin is an abundant scaffold protein in the postsynaptic density (PSD) that forms a high-affinity complex with αCaMKII and α-actinin. To assess the function of densin, we created a mouse line with a null mutation in the gene encoding it (LRRC7). Homozygous knock-out mice display a wide variety of abnormal behaviors that are often considered endophenotypes of schizophrenia and autism spectrum disorders. At the cellular level, loss of densin results in reduced levels of α-actinin in the brain and selective reduction in the localization of mGluR5 and DISC1 in the PSD fraction, whereas the amounts of ionotropic glutamate receptors and other prominent PSD proteins are unchanged. In addition, deletion of densin results in impairment of mGluR- and NMDA receptor-dependent forms of long-term depression, alters the early dynamics of regulation of CaMKII by NMDA-type glutamate receptors, and produces a change in spine morphology. These results indicate that densin influences the function of mGluRs and CaMKII at synapses and contributes to localization of mGluR5 and DISC1 in the PSD fraction. They are consistent with the hypothesis that mutations that disrupt the organization and/or dynamics of postsynaptic signaling complexes in excitatory synapses can cause behavioral endophenotypes of mental illness.


Subject(s)
Gene Expression Regulation/genetics , Mental Disorders/genetics , Nerve Tissue Proteins/metabolism , Post-Synaptic Density/metabolism , Receptors, Kainic Acid/metabolism , Sialoglycoproteins/deficiency , Actins/metabolism , Aggression/physiology , Animals , Behavior, Animal/physiology , Bicuculline/pharmacology , Body Weight/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cells, Cultured , Dendritic Spines/metabolism , Disease Models, Animal , Embryo, Mammalian , Endophenotypes , Exploratory Behavior/physiology , Female , GABA Antagonists/pharmacology , GABA-A Receptor Antagonists/pharmacology , Genotype , Glial Fibrillary Acidic Protein/metabolism , Glycine/pharmacology , Green Fluorescent Proteins/genetics , Hippocampus/cytology , In Vitro Techniques , Inhibition, Psychological , Long-Term Potentiation/genetics , Long-Term Synaptic Depression/genetics , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Memory, Short-Term/physiology , Mental Disorders/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/genetics , Muscle Strength/genetics , Mutation/genetics , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/drug effects , Patch-Clamp Techniques , Picrotoxin/pharmacology , Psychomotor Performance/physiology , Receptors, AMPA/genetics , Receptors, Kainic Acid/genetics , Recognition, Psychology/physiology , Rotarod Performance Test , Statistics, Nonparametric , Time Factors
9.
J Vis Exp ; (49)2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21445033

ABSTRACT

Brain injury, genetic manipulations, and pharmacological treatments can result in alterations of motor skills in mice. Fine motor coordination and balance can be assessed by the beam walking assay. The goal of this test is for the mouse to stay upright and walk across an elevated narrow beam to a safe platform. This test takes place over 3 consecutive days: 2 days of training and 1 day of testing. Performance on the beam is quantified by measuring the time it takes for the mouse to traverse the beam and the number of paw slips that occur in the process. Here we report the protocol used in our laboratory, and representative results from a cohort of C57BL/6 mice. This task is particularly useful for detecting subtle deficits in motor skills and balance that may not be detected by other motor tests, such as the Rotarod.


Subject(s)
Motor Skills/physiology , Postural Balance/physiology , Psychomotor Performance/physiology , Task Performance and Analysis , Animals , Female , Male , Mice , Mice, Inbred C57BL , Rotarod Performance Test
10.
EMBO Rep ; 11(6): 473-8, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20467438

ABSTRACT

Sensitization to inflammatory pain is a pathological form of neuronal plasticity that is poorly understood and treated. Here we examine the role of the SH3 domain of postsynaptic density 95 (PSD95) by using mice that carry a single amino-acid substitution in the polyproline-binding site. Testing multiple forms of plasticity we found sensitization to inflammation was specifically attenuated. The inflammatory response required recruitment of phosphatidylinositol-3-kinase-C2alpha to the SH3-binding site of PSD95. In wild-type mice, wortmannin or peptide competition attenuated the sensitization. These results show that different types of behavioural plasticity are mediated by specific domains of PSD95 and suggest novel therapeutic avenues for reducing inflammatory pain.


Subject(s)
Inflammation/complications , Inflammation/enzymology , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Pain/complications , Pain/enzymology , Phosphatidylinositol 3-Kinases/metabolism , src Homology Domains , Animals , Disks Large Homolog 4 Protein , Guanylate Kinases , Hippocampus/enzymology , Hippocampus/pathology , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Neuronal Plasticity , Point Mutation/genetics , Protein Binding , Structure-Activity Relationship , Synapses/enzymology
11.
J Neurosci ; 28(50): 13673-83, 2008 Dec 10.
Article in English | MEDLINE | ID: mdl-19074040

ABSTRACT

SynGAP, a prominent Ras/Rap GTPase-activating protein in the postsynaptic density, regulates the timing of spine formation and trafficking of glutamate receptors in cultured neurons. However, the molecular mechanisms by which it does this are unknown. Here, we show that synGAP is a key regulator of spine morphology in adult mice. Heterozygous deletion of synGAP was sufficient to cause an excess of mushroom spines in adult brains, indicating that synGAP is involved in steady-state regulation of actin in mature spines. Both Ras- and Rac-GTP levels were elevated in forebrains from adult synGAP(+/-) mice. Rac is a well known regulator of actin polymerization and spine morphology. The steady-state level of phosphorylation of cofilin was also elevated in synGAP(+/-) mice. Cofilin, an F-actin severing protein that is inactivated by phosphorylation, is a downstream target of a pathway regulated by Rac. We show that transient regulation of cofilin by treatment with NMDA is also disrupted in synGAP mutant neurons. Treatment of wild-type neurons with 25 mum NMDA triggered transient dephosphorylation and activation of cofilin within 15 s. In contrast, neurons cultured from mice with a homozygous or heterozygous deletion of synGAP lacked the transient regulation by the NMDA receptor. Depression of EPSPs induced by a similar treatment of hippocampal slices with NMDA was disrupted in slices from synGAP(+/-) mice. Our data show that synGAP mediates a rate-limiting step in steady-state regulation of spine morphology and in transient NMDA-receptor-dependent regulation of the spine cytoskeleton.


Subject(s)
Actin Depolymerizing Factors/metabolism , Dendritic Spines/metabolism , Dendritic Spines/ultrastructure , ras GTPase-Activating Proteins/metabolism , Animals , Blotting, Western , Brain/metabolism , Brain/ultrastructure , Cytoskeleton , Dendritic Spines/drug effects , Electrophoresis, Polyacrylamide Gel , Excitatory Amino Acid Agonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Long-Term Synaptic Depression/drug effects , Long-Term Synaptic Depression/physiology , Mice , Mice, Knockout , Microscopy, Confocal , N-Methylaspartate/pharmacology , Organ Culture Techniques , Patch-Clamp Techniques , Phosphorylation , Receptors, N-Methyl-D-Aspartate/metabolism , rac GTP-Binding Proteins/metabolism , ras GTPase-Activating Proteins/genetics , ras Proteins/metabolism
12.
J Physiol ; 586(24): 5885-900, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18936077

ABSTRACT

The membrane-associated guanylate kinases (MAGUKs) PSD-95, PSD-93 and SAP102 are thought to have crucial roles in both AMPA receptor trafficking and formation of NMDA receptor-associated signalling complexes involved in synaptic plasticity. While PSD-95, PSD-93, and SAP102 appear to have similar roles in AMPA receptor trafficking, it is not known whether these MAGUKs also have functionally similar roles in synaptic plasticity. To explore this issue we examined several properties of basal synaptic transmission in the hippocampal CA1 region of PSD-93 and PSD-95 mutant mice and compared the ability of a number of different synaptic stimulation protocols to induce long-term potentiation (LTP) and long-term depression (LTD) in these mutants. We find that while both AMPA and NMDA receptor-mediated synaptic transmission are normal in PSD-93 mutants, PSD-95 mutant mice exhibit clear deficits in AMPA receptor-mediated transmission. Moreover, in contrast to the facilitation of LTP induction and disruption of LTD observed in PSD-95 mutant mice, PSD-93 mutant mice exhibit deficits in LTP and normal LTD. Our results suggest that PSD-95 has a unique role in AMPA receptor trafficking at excitatory synapses in the hippocampus of adult mice and indicate that PSD-93 and PSD-95 have essentially opposite roles in LTP, perhaps because these MAGUKs form distinct NMDA receptor signalling complexes that differentially regulate the induction of LTP by different patterns of synaptic activity.


Subject(s)
Action Potentials/physiology , Intracellular Signaling Peptides and Proteins/physiology , Long-Term Potentiation/physiology , Membrane Proteins/physiology , Neuronal Plasticity/physiology , Animals , Disks Large Homolog 4 Protein , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Guanylate Kinases , Hippocampus/physiology , Intracellular Signaling Peptides and Proteins/genetics , Lidocaine/analogs & derivatives , Lidocaine/pharmacology , Long-Term Potentiation/drug effects , Long-Term Synaptic Depression/physiology , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Patch-Clamp Techniques , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Receptors, AMPA/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Sodium Channel Blockers/pharmacology , Synaptic Transmission/physiology
13.
Nat Rev Neurosci ; 6(6): 423-34, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15928715

ABSTRACT

Short-term and long-term changes in the strength of synapses in neural networks underlie working memory and long-term memory storage in the brain. These changes are regulated by many biochemical signalling pathways in the postsynaptic spines of excitatory synapses. Recent findings about the roles and regulation of the small GTPases Ras, Rap and Rac in spines provide new insights into the coordination and cooperation of different pathways to effect synaptic plasticity. Here, we present an initial working representation of the interactions of five signalling cascades that are usually studied individually. We discuss their integrated function in the regulation of postsynaptic plasticity.


Subject(s)
Dendritic Spines/physiology , Neuronal Plasticity/physiology , Signal Transduction/physiology , Synapses/physiology , Animals , Calcium Signaling/physiology , Dendritic Spines/chemistry , Dendritic Spines/metabolism , Humans , Synapses/chemistry , Synapses/metabolism
14.
Trends Neurosci ; 28(4): 182-7, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15808352

ABSTRACT

Many forms of mental retardation and cognitive disability are associated with abnormalities in dendritic spine morphology. Visualization of spines using live-imaging techniques provides convincing evidence that spine morphology is altered in response to certain forms of LTP-inducing stimulation. Thus, information storage at the cellular level appears to involve changes in spine morphology that support changes in synaptic strength produced by certain patterns of synaptic activity. Because the structure of a spine is determined by its underlying actin cytoskeleton, there has been much effort to identify signaling pathways linking synaptic activity to control of actin polymerization. This review, part of the TINS Synaptic Connectivity series, discusses recent studies that implicate EphB and NMDA receptors in the regulation of actin-binding proteins through modulation of Rho family small GTPases.


Subject(s)
Dendritic Spines/physiology , Neuronal Plasticity/physiology , Synapses/physiology , Animals , Cytoskeletal Proteins/physiology , Humans , Long-Term Potentiation/physiology , Models, Neurological
15.
J Neurosci ; 22(22): 9721-32, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12427827

ABSTRACT

At excitatory synapses, the postsynaptic scaffolding protein postsynaptic density 95 (PSD-95) couples NMDA receptors (NMDARs) to the Ras GTPase-activating protein SynGAP. The close association of SynGAP and NMDARs suggests that SynGAP may have an important role in NMDAR-dependent activation of Ras signaling pathways, such as the MAP kinase pathway, and in synaptic plasticity. To explore this issue, we examined long-term potentiation (LTP), p42 MAPK (ERK2) signaling, and spatial learning in mice with a heterozygous null mutation of the SynGAP gene (SynGAP(-/+)). In SynGAP(-/+) mutant mice, the induction of LTP in the hippocampal CA1 region was strongly reduced in the absence of any detectable alteration in basal synaptic transmission and NMDAR-mediated synaptic currents. Although basal levels of activated ERK2 were elevated in hippocampal extracts from SynGAP(-/+) mice, NMDAR stimulation still induced a robust increase in ERK activation in slices from SynGAP(-/+) mice. Thus, although SynGAP may regulate the ERK pathway, its role in LTP most likely involves additional downstream targets. Consistent with this, the amount of potentiation induced by stimulation protocols that induce an ERK-independent form of LTP were also significantly reduced in slices from SynGAP(-/+) mice. An elevation of basal phospho-ERK2 levels and LTP deficits were also observed in SynGAP(-/+)/H-Ras(-)/- double mutants, suggesting that SynGAP may normally regulate Ras isoforms other than H-Ras. A comparison of SynGAP and PSD-95 mutants suggests that PSD-95 couples NMDARs to multiple downstream signaling pathways with very different roles in LTP and learning.


Subject(s)
GTPase-Activating Proteins/metabolism , Learning/physiology , MAP Kinase Signaling System/physiology , Neuronal Plasticity/physiology , Synapses/metabolism , Animals , Disks Large Homolog 4 Protein , Electric Stimulation , Excitatory Postsynaptic Potentials/physiology , Fetal Viability/genetics , GTPase-Activating Proteins/genetics , Gene Targeting , Guanylate Kinases , Heterozygote , Hippocampus/chemistry , Hippocampus/cytology , Hippocampus/metabolism , In Vitro Techniques , Intracellular Signaling Peptides and Proteins , Long-Term Potentiation/physiology , Macromolecular Substances , Membrane Proteins , Mice , Mice, Mutant Strains , Mitogen-Activated Protein Kinase 1/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Patch-Clamp Techniques , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission/physiology , ras Proteins/genetics , ras Proteins/metabolism
16.
J Neurophysiol ; 87(3): 1395-403, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11877514

ABSTRACT

Activation of metabotropic glutamate receptors (mGluRs) with the group I mGluR selective agonist (R,S)-3,5-dihydroxyphenylglycine (DHPG) induces a long-term depression (LTD) of excitatory synaptic transmission in the CA1 region of the hippocampus. Here we investigated the potential roles of pre- and postsynaptic processes in the DHPG-induced LTD at excitatory synapses onto hippocampal pyramidal cells in the mouse hippocampus. Activation of mGluRs with DHPG, but not ACPD, induced LTD at both Schaffer collateral/commissural fiber synapses onto CA1 pyramidal cells and at associational/commissural fiber synapses onto CA3 pyramidal cells. DHPG-induced LTD was blocked when the G-protein inhibitor guanosine-5'-O-(2-thiodiphosphate) was selectively delivered into postsynaptic CA1 pyramidal cells via an intracellular recording electrode, suggesting that DHPG depresses synaptic transmission through a postsynaptic, GTP-dependent signaling pathway. The effects of DHPG were also strongly modulated, however, by experimental manipulations that altered presynaptic calcium influx. In these experiments, we found that elevating extracellular Ca(2+) concentrations ([Ca(2+)](o)) to 6 mM almost completely blocked the effects of DHPG, whereas lowering [Ca(2+)](o) to 1 mM significantly enhanced the ability of DHPG to depress synaptic transmission. Enhancing Ca(2+) influx by prolonging action potential duration with bath applications of the K(+) channel blocker 4-aminopyridine (4-AP) also strongly reduced the effects of DHPG in the presence of normal [Ca(2+)](o) (2 mM). Although these findings indicate that alterations in Ca(2+)-dependent signaling processes strongly regulate the effects of DHPG on synaptic transmission, they do not distinguish between potential pre- versus postsynaptic sites of action. We found, however, that while inhibiting both pre- and postsynaptic K(+) channels with bath-applied 4-AP blocked the effects of DHPG; inhibition of postsynaptic K(+) channels alone with intracellular Cs(+) and TEA had no effect on the ability of DHPG to inhibit synaptic transmission. This suggests that presynaptic changes in transmitter release contribute to the depression of synaptic transmission by DHPG. Consistent with this, DHPG induced a persistent depression of both AMPA and N-methyl-D-aspartate receptor-mediated components of excitatory postsynaptic currents in voltage-clamped pyramidal cells. Together our results suggest that activation of postsynaptic mGluRs suppresses transmission at excitatory synapses onto CA1 pyramidal cells through presynaptic effects on transmitter release.


Subject(s)
Hippocampus/physiology , Methoxyhydroxyphenylglycol/analogs & derivatives , Neural Inhibition/physiology , Receptors, Metabotropic Glutamate/physiology , 4-Aminopyridine/pharmacology , Amino Acids/pharmacology , Animals , Calcium Signaling/physiology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Methoxyhydroxyphenylglycol/pharmacology , Mice , Mice, Inbred C57BL , Neural Inhibition/drug effects , Organ Culture Techniques , Potassium Channel Blockers/pharmacology , Presynaptic Terminals/physiology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Xanthenes/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...