Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
PNAS Nexus ; 3(2): pgae062, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38384385

ABSTRACT

Metabotropic glutamate receptor-dependent long-term depression (mGluR-LTD) is an important form of synaptic plasticity that occurs in many regions of the central nervous system and is the underlying mechanism for several learning paradigms. In the hippocampus, mGluR-LTD is manifested by the weakening of synaptic transmission and elimination of dendritic spines. Interestingly, not all spines respond or undergo plasticity equally in response to mGluR-LTD. A subset of dendritic spines containing synaptopodin (SP), an actin-associated protein is critical for mGluR-LTD and protects spines from elimination through mGluR1 activity. The precise cellular function of SP is still enigmatic and it is still unclear how SP contributes to the functional aspect of mGluR-LTD despite its modulation of the structural plasticity. In this study, we show that the lack of SP impairs mGluR-LTD by negatively affecting the mGluR5-dependent activity. Such impairment of mGluR5 activity is accompanied by a significant decrease of surface mGluR5 level in SP knockout (SPKO) mice. Intriguingly, the remaining mGluR-LTD becomes a protein synthesis-independent process in the SPKO and is mediated instead by endocannabinoid signaling. These data indicate that the postsynaptic protein SP can regulate the locus of expression of mGluR-LTD and provide insight into our understanding of spine/synapse-specific plasticity.

2.
bioRxiv ; 2023 Aug 03.
Article in English | MEDLINE | ID: mdl-37577654

ABSTRACT

Metabotropic glutamate receptor-dependent long-term depression (mGluR-LTD) is an important form of synaptic plasticity that occurs in many regions of the CNS and is the underlying mechanism for several learning paradigms. In the hippocampus, mGluR-LTD is manifested by the weakening of synaptic transmission and elimination of dendritic spines. Interestingly, not all spines respond or undergo plasticity equally in response to mGluR-LTD. A subset of dendritic spines containing synaptopodin (SP), an actin-associated protein, are critical for mGluR-LTD and protect spines from elimination through mGluR1 activity. The precise cellular function of SP is still enigmatic and it is still unclear how SP contributes to the functional aspect of mGluR-LTD despite of its modulation on the structural plasticity. In the present study, we show that the lack of SP impairs mGluR-LTD by negatively affecting the mGluR5-dependent activity. Such impairment of mGluR5 activity is accompanied by a significant decrease of surface mGluR5 level in SP knockout (SPKO) mice. Intriguingly, the remaining mGluR-LTD becomes a protein synthesis-independent process in the SPKO and is mediated instead by endocannabinoid signaling. These data show for the first time that the postsynaptic protein SP can regulate the locus of expression of mGluR-LTD and provide insight to our understanding of spine/synapse-specific plasticity. Significance statement: Hippocampal group I metabotropic glutamate receptor dependent long-term depression (mGluR-LTD), a form of learning and memory, is misregulated in many murine models of neurodevelopmental disorders. Despite extensive studies there is a paucity of information on the molecular mechanism underlying mGluR-LTD. Previously, we reported that loss of synaptopodin, an actin-associated protein found in a subset of mature dendritic spines, impairs mGluR-LTD. In the current study, we uncover the molecular and cellular deficits involved. We find that synaptopodin is required for the mGluR5-Homer interaction and uncover synaptopodin as a molecular switch for mGluR-LTD expression, as mGluR-LTD becomes protein synthesis-independent and relies on endocannabinoid signaling in synaptopodin knock-out. This work provides insight into synaptopodin as a gatekeeper to regulate mGluR-LTD at hippocampal synapses.

3.
Biomedicines ; 10(11)2022 Oct 25.
Article in English | MEDLINE | ID: mdl-36359212

ABSTRACT

Structural, functional, and molecular alterations in excitatory spines are a common hallmark of many neurodevelopmental disorders including intellectual disability and autism. Here, we describe an optimized methodology, based on combined use of DiI and immunofluorescence, for rapid and sensitive characterization of the structure and composition of spines in native brain tissue. We successfully demonstrate the applicability of this approach by examining the properties of hippocampal spines in juvenile Fmr1 KO mice, a mouse model of Fragile X Syndrome. We find that mutant mice display pervasive dysgenesis of spines evidenced by an overabundance of both abnormally elongated thin spines and cup-shaped spines, in combination with reduced density of mushroom spines. We further find that mushroom spines expressing the actin-binding protein Synaptopodin-a marker for spine apparatus-are more prevalent in mutant mice. Previous work identified spines with Synaptopodin/spine apparatus as the locus of mGluR-LTD, which is abnormally elevated in Fmr1 KO mice. Altogether, our data suggest this enhancement may be linked to the preponderance of this subset of spines in the mutant. Overall, these findings demonstrate the sensitivity and versatility of the optimized methodology by uncovering a novel facet of spine dysgenesis in Fmr1 KO mice.

4.
J Neurosci ; 42(9): 1666-1678, 2022 03 02.
Article in English | MEDLINE | ID: mdl-35046120

ABSTRACT

Dendritic spines, actin-rich protrusions forming the postsynaptic sites of excitatory synapses, undergo activity-dependent molecular and structural remodeling. Activation of Group 1 metabotropic glutamate receptors (mGluR1 and mGluR5) by synaptic or pharmacological stimulation, induces LTD, but whether this is accompanied with spine elimination remains unresolved. A subset of telencephalic mushroom spines contains the spine apparatus (SA), an enigmatic organelle composed of stacks of smooth endoplasmic reticulum, whose formation depends on the expression of the actin-bundling protein Synaptopodin. Allocation of Synaptopodin to spines appears governed by cell-intrinsic mechanisms as the relative frequency of spines harboring Synaptopodin is conserved in vivo and in vitro Here we show that expression of Synaptopodin/SA in spines is required for induction of mGluR-LTD at Schaffer collateral-CA1 synapses of male mice. Post-mGluR-LTD, mushroom spines lacking Synaptopodin/SA are selectively lost, whereas spines harboring it are preserved. This process, dependent on activation of mGluR1 but not mGluR5, is conserved in mature mouse neurons and rat neurons of both sexes. Mechanistically, we find that mGluR1 supports physical retention of Synaptopodin within excitatory spine synapses during LTD while triggering lysosome-dependent degradation of the protein residing in dendritic shafts. Together, these results reveal a cellular mechanism, dependent on mGluR1, which enables selective preservation of stronger spines containing Synaptopodin/SA while eliminating weaker ones and potentially countering spurious strengthening by de novo recruitment of Synaptopodin. Overall, our results identify spines with Synaptopodin/SA as the locus of mGluR-LTD and underscore the importance of the molecular microanatomy of spines in synaptic plasticity.SIGNIFICANCE STATEMENT Long-term changes in functional synaptic strength are associated with modification of synaptic connectivity through stabilization or elimination of dendritic spines, the postsynaptic locus of excitatory synapses. How heterogeneous spine microanatomy instructs spine remodeling after long-term synaptic depression (LTD) remains unclear. Metabotropic glutamate receptors mGluR1 and mGluR5 induce a form of LTD critical to circuit function in physiological and disease conditions. Our results identify spines containing the protein Synaptopodin, which enables local assembly of a spine apparatus, as the locus of expression of mGluR-LTD and demonstrate a specific role of mGluR1 in promoting selective loss after mGluR-LTD of mature dendritic spines lacking Synaptopodin/spine apparatus. These findings highlight the fundamental contribution of spine microanatomy in selectively enabling functional and structural plasticity.


Subject(s)
Actins , Long-Term Synaptic Depression , Receptors, Metabotropic Glutamate , Synapses , Actins/metabolism , Animals , Dendritic Spines/metabolism , Female , Hippocampus/metabolism , Hippocampus/physiology , Long-Term Synaptic Depression/physiology , Male , Mice , Neuronal Plasticity/physiology , Rats , Receptors, Metabotropic Glutamate/metabolism , Synapses/physiology
5.
Curr Neuropharmacol ; 14(5): 494-503, 2016.
Article in English | MEDLINE | ID: mdl-27296642

ABSTRACT

Group I metabotropic glutamate receptors mediate slow excitatory neurotransmission in the central nervous system and are critical to activity-dependent synaptic plasticity, a cellular substrate of learning and memory. Dysregulated receptor signaling is implicated in neuropsychiatric conditions ranging from neurodevelopmental to neurodegenerative disorders. Importantly, group I metabotropic glutamate receptor signaling functions can be modulated by interacting proteins that mediate receptor trafficking, expression and coupling efficiency to signaling effectors. These interactions afford cell- or pathway-specific modulation to fine-tune receptor function, thus representing a potential target for pharmacological interventions in pathological conditions.


Subject(s)
Inflammation/metabolism , Neurodegenerative Diseases/metabolism , Neuronal Plasticity/physiology , Pain/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Humans
6.
Neuron ; 90(6): 1189-1202, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27263971

ABSTRACT

Circuit function in the CNS relies on the balanced interplay of excitatory and inhibitory synaptic signaling. How neuronal activity influences synaptic differentiation to maintain such balance remains unclear. In the mouse spinal cord, a population of GABAergic interneurons, GABApre, forms synapses with the terminals of proprioceptive sensory neurons and controls information transfer at sensory-motor connections through presynaptic inhibition. We show that reducing sensory glutamate release results in decreased expression of GABA-synthesizing enzymes GAD65 and GAD67 in GABApre terminals and decreased presynaptic inhibition. Glutamate directs GAD67 expression via the metabotropic glutamate receptor mGluR1ß on GABApre terminals and regulates GAD65 expression via autocrine influence on sensory terminal BDNF. We demonstrate that dual retrograde signals from sensory terminals operate hierarchically to direct the molecular differentiation of GABApre terminals and the efficacy of presynaptic inhibition. These retrograde signals comprise a feedback mechanism by which excitatory sensory activity drives GABAergic inhibition to maintain circuit homeostasis.


Subject(s)
Glutamic Acid/physiology , Neural Inhibition/physiology , Neurons/physiology , Presynaptic Terminals/physiology , Receptors, Metabotropic Glutamate/physiology , Synapses/physiology , Animals , Brain-Derived Neurotrophic Factor/physiology , Glutamate Decarboxylase/biosynthesis , Glutamic Acid/metabolism , Interneurons/physiology , Mice , Models, Neurological , Neurons/metabolism , Presynaptic Terminals/metabolism , Sensory Receptor Cells/metabolism , Spinal Cord/metabolism , Spinal Cord/physiology , Synapses/metabolism , Vesicular Glutamate Transport Protein 1/genetics , gamma-Aminobutyric Acid/biosynthesis
7.
J Biol Chem ; 290(26): 15909-20, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-25944910

ABSTRACT

Dendritic spines are dynamic, actin-rich protrusions in neurons that undergo remodeling during neuronal development and activity-dependent plasticity within the central nervous system. Although group 1 metabotropic glutamate receptors (mGluRs) are critical for spine remodeling under physiopathological conditions, the molecular components linking receptor activity to structural plasticity remain unknown. Here we identify a Ca(2+)-sensitive actin-binding protein, α-actinin-4, as a novel group 1 mGluR-interacting partner that orchestrates spine dynamics and morphogenesis in primary neurons. Functional silencing of α-actinin-4 abolished spine elongation and turnover stimulated by group 1 mGluRs despite intact surface receptor expression and downstream ERK1/2 signaling. This function of α-actinin-4 in spine dynamics was underscored by gain-of-function phenotypes in untreated neurons. Here α-actinin-4 induced spine head enlargement, a morphological change requiring the C-terminal domain of α-actinin-4 that binds to CaMKII, an interaction we showed to be regulated by group 1 mGluR activation. Our data provide mechanistic insights into spine remodeling by metabotropic signaling and identify α-actinin-4 as a critical effector of structural plasticity within neurons.


Subject(s)
Actinin/metabolism , Dendritic Spines/metabolism , Receptors, Metabotropic Glutamate/metabolism , Actinin/genetics , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cells, Cultured , Dendritic Spines/genetics , Female , Humans , Male , Mice , Neurons/cytology , Neurons/metabolism , Protein Binding , Receptors, Metabotropic Glutamate/genetics
8.
PLoS One ; 10(4): e0121464, 2015.
Article in English | MEDLINE | ID: mdl-25849048

ABSTRACT

Fragile X Syndrome, a leading cause of inherited intellectual disability and autism, arises from transcriptional silencing of the FMR1 gene encoding an RNA-binding protein, Fragile X Mental Retardation Protein (FMRP). FMRP can regulate the expression of approximately 4% of brain transcripts through its role in regulation of mRNA transport, stability and translation, thus providing a molecular rationale for its potential pleiotropic effects on neuronal and brain circuitry function. Several intracellular signaling pathways are dysregulated in the absence of FMRP suggesting that cellular deficits may be broad and could result in homeostatic changes. Lipid rafts are specialized regions of the plasma membrane, enriched in cholesterol and glycosphingolipids, involved in regulation of intracellular signaling. Among transcripts targeted by FMRP, a subset encodes proteins involved in lipid biosynthesis and homeostasis, dysregulation of which could affect the integrity and function of lipid rafts. Using a quantitative mass spectrometry-based approach we analyzed the lipid raft proteome of Fmr1 knockout mice, an animal model of Fragile X syndrome, and identified candidate proteins that are differentially represented in Fmr1 knockout mice lipid rafts. Furthermore, network analysis of these candidate proteins reveals connectivity between them and predicts functional connectivity with genes encoding components of myelin sheath, axonal processes and growth cones. Our findings provide insight to aid identification of molecular and cellular dysfunctions arising from Fmr1 silencing and for uncovering shared pathologies between Fragile X syndrome and other autism spectrum disorders.


Subject(s)
Brain/metabolism , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/metabolism , Membrane Microdomains/metabolism , Membrane Proteins/metabolism , Proteome/metabolism , Animals , Brain/pathology , Disease Models, Animal , Fragile X Syndrome/genetics , Fragile X Syndrome/pathology , Humans , Membrane Microdomains/genetics , Membrane Microdomains/pathology , Membrane Proteins/genetics , Mice , Mice, Knockout , Proteome/genetics
9.
J Biol Chem ; 288(44): 32004-19, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24045944

ABSTRACT

Group I metabotropic glutamate receptors (mGluRs), mGluR1 and mGluR5, play critical functions in forms of activity-dependent synaptic plasticity and synapse remodeling in physiological and pathological states. Importantly, in animal models of fragile X syndrome, group I mGluR activity is abnormally enhanced, a dysfunction that may partly underlie cognitive deficits in the condition. Lipid rafts are cholesterol- and sphingolipid-enriched membrane domains that are thought to form transient signaling platforms for ligand-activated receptors. Many G protein-coupled receptors, including group I mGluRs, are present in lipid rafts, but the mechanisms underlying recruitment to these membrane domains remain incompletely understood. Here, we show that mGluR1 recruitment to lipid rafts is enhanced by agonist binding and is supported at least in part by an intact cholesterol recognition/interaction amino acid consensus (CRAC) motif in the receptor. Substitutions of critical residues in the motif reduce mGluR1 association with lipid rafts and agonist-induced, mGluR1-dependent activation of extracellular-signal-activated kinase1/2 MAP kinase (ERK-MAPK). We find that alteration of membrane cholesterol content or perturbation of lipid rafts regulates agonist-dependent activation of ERK-MAPK by group I mGluRs, suggesting a potential function for cholesterol as a positive allosteric modulator of receptor function(s). Together, these findings suggest that drugs that alter membrane cholesterol levels or directed to the receptor-cholesterol interface could be employed to modulate abnormal group I mGluR activity in neuropsychiatric conditions, including fragile X syndrome.


Subject(s)
Cholesterol/metabolism , Fragile X Syndrome/metabolism , MAP Kinase Signaling System , Membrane Microdomains/metabolism , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/metabolism , Allosteric Regulation/genetics , Amino Acid Motifs , Animals , Cholesterol/genetics , Fragile X Syndrome/genetics , HEK293 Cells , Humans , Membrane Microdomains/genetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Protein Structure, Tertiary , Receptors, Metabotropic Glutamate/genetics
10.
Methods Mol Biol ; 746: 411-23, 2011.
Article in English | MEDLINE | ID: mdl-21607872

ABSTRACT

Lipid domains of the plasma membrane were originally described as a cell matrix insoluble in cold -nonionic detergents and enriched in glycosphingolipids. Because of these biochemical properties, these membrane domains were termed detergent-resistant membranes (DRMs) or detergent-insoluble -glycolipid-enriched (DIG) membranes. Membrane rafts and caveolae are two types of lipid domains that share these properties, as well as structural/functional dependence on membrane cholesterol. Membrane rafts and caveolae are believed to act as signaling platforms for ligand-activated receptors, thereby contributing to the regulation of receptor function. Here we describe a simple method to assess the association of GPCRs with detergent resistant membranes in native brain tissue and cultured cells.


Subject(s)
Cell Membrane/drug effects , Cell Membrane/metabolism , Detergents/pharmacology , Receptors, G-Protein-Coupled/metabolism , Animals , Biological Assay , Brain/metabolism , HEK293 Cells , Humans , Male , Protein Transport , Subcellular Fractions
11.
Proc Natl Acad Sci U S A ; 107(50): 21778-83, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21098662

ABSTRACT

Group I metabotropic glutamate receptors (mGluR1/5) are important to synaptic circuitry formation during development and to forms of activity-dependent synaptic plasticity. Dysregulation of mGluR1/5 signaling is implicated in some disorders of neurodevelopment, including fragile X syndrome, the most common inherited form of intellectual disabilities and leading cause of autism. Site(s) in the intracellular loops of mGluR1/5 directly bind caveolin-1, an adaptor protein that associates with membrane rafts. Caveolin-1 is the main coat component of caveolae and organizes macromolecular signaling complexes with effector proteins and membrane receptors. We report that long-term depression (LTD) elicited by a single application of the group I mGluR selective agonist (RS)-3,5-dihydroxyphenylglycine (DHPG) was markedly attenuated at Schaffer collateral-CA1 synapses of mice lacking caveolin-1 (Cav1(-/-)), as assessed by field recording. In contrast, multiple applications of DHPG produced LTD comparable to that in WT mice. Passive membrane properties, basal glutamatergic transmission and NMDA receptor (NMDAR)-dependent LTD were unaltered. The remaining LTD was reduced by anisomycin, an inhibitor of protein synthesis, by U0126, an inhibitor of MEK1/2 kinases, and by rapamycin, an inhibitor of mammalian target of rapamycin (mTOR), suggesting mediation by the same mechanisms as in WT. mGluR1/5-dependent activation (phosphorylation) of MEK and extracellular signal-regulated kinase (ERK1/2) was altered in Cav1(-/-) mice; basal phosphorylation was increased, but a single application of DHPG had no further effect, and after DHPG, phosphorylation was similar in WT and Cav1(-/-) mice. Taken together, our findings suggest that caveolin-1 is required for normal coupling of mGluR1/5 to downstream signaling cascades and induction of mGluR-LTD.


Subject(s)
Caveolin 1/metabolism , Hippocampus/cytology , Hippocampus/physiology , Long-Term Synaptic Depression/physiology , Receptors, Metabotropic Glutamate/metabolism , Synapses/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Butadienes/pharmacology , Caveolin 1/genetics , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycine/analogs & derivatives , Glycine/pharmacology , Long-Term Synaptic Depression/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitriles/pharmacology , Patch-Clamp Techniques , Resorcinols/pharmacology , Signal Transduction/physiology , Sirolimus/pharmacology , Synapses/drug effects , TOR Serine-Threonine Kinases/metabolism
12.
Pacing Clin Electrophysiol ; 33(7): 841-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20180916

ABSTRACT

BACKGROUND: Few studies have assessed the long-term effects of cardiac resynchronization therapy (CRT) in patients with advanced heart failure (HF) and previously right ventricular apical pacing (RVAP). AIMS: To assess the clinical and hemodynamic impact of upgrading to biventricular pacing in patients with severe HF and permanent RVAP in comparison with patients who had CRT implantation as initial therapy. METHODS AND RESULTS: Thirty-nine patients with RVAP, advanced HF (New York Heart Association [NYHA] III-IV), and severe depression of left ventricular ejection fraction (LVEF) were upgraded to biventricular pacing (group A). Mean duration of RVAP before upgrading was 41.8 +/- 13.3 months. Clinical and echocardiographic results were compared to those obtained in a group of 43 patients with left bundle branch block and similar clinical characteristics undergoing "primary" CRT (group B). Mean follow-up was 35 +/- 10 months in patients of group A and 38 +/- 12 months in group B. NYHA class significantly improved in groups A and B. LVEF increased from 0.23 +/- 0.07 to 0.36 +/- 0.09 (P < 0.001) and from 0.26 +/- 0.02 to 0.34 +/- 0.10 (P < 0.001), respectively. Hospitalizations were reduced by 81% and 77% (P < 0.001). Similar improvements in echocardiographic signs of ventricular desynchronization were also observed. CONCLUSION: Patients upgraded to CRT exhibit long-term clinical and hemodynamic benefits that are similar to those observed in patients treated with CRT as initial strategy.


Subject(s)
Cardiac Pacing, Artificial/methods , Heart Failure/diagnosis , Heart Failure/prevention & control , Pacemaker, Artificial , Ventricular Dysfunction, Left/diagnosis , Ventricular Dysfunction, Left/prevention & control , Aged , Equipment Design , Equipment Failure Analysis , Female , Heart Failure/complications , Humans , Male , Reproducibility of Results , Sensitivity and Specificity , Treatment Outcome , Ventricular Dysfunction, Left/etiology
13.
Europace ; 11(9): 1227-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19549675

ABSTRACT

We report the case of a 59-year-old non-Caucasian man with sustained left ventricular (LV) tachycardia and isolated LV non-compaction. An electro-anatomical mapping of the right ventricle and LV with the Carto system was reconstructed. The voltage map excluded the presence of scarred tissue as a possible substrate responsible of the ventricular arrhythmia.


Subject(s)
Body Surface Potential Mapping/methods , Cardiomyopathies/complications , Cardiomyopathies/diagnosis , Tachycardia, Ventricular/complications , Tachycardia, Ventricular/diagnosis , Ventricular Dysfunction, Left/complications , Ventricular Dysfunction, Left/diagnosis , Diagnosis, Differential , Humans , Male , Middle Aged
14.
J Neurosci ; 29(11): 3590-602, 2009 Mar 18.
Article in English | MEDLINE | ID: mdl-19295163

ABSTRACT

Endocytic trafficking of neurotransmitter receptors is critical to neuronal signaling and activity-dependent synaptic plasticity. Although the importance of clathrin-mediated endocytosis in receptor trafficking in neurons is well established, the contribution of the caveolar/lipid raft pathway has been little explored. Here, we show that caveolin-1, an adaptor protein that associates with lipid rafts and the main coat protein of caveolae, binds to and colocalizes with metabotropic glutamate receptors 1/5 (mGluR1/5). The interaction with caveolin-1 controls the rate of constitutive mGluR1 internalization, thereby regulating expression of the receptor at the cell surface. Consistent with a role for caveolin-1 in mGluR trafficking, we show that mGluR1/5 associate with lipid rafts in the brain and that their constitutive internalization is mediated, in both heterologous cells and neurons, by caveolar/raft-dependent endocytosis. We further show that caveolin-1 attenuates mGluR1-dependent activation of extracellular signal-regulated kinase (ERK)-mitogen-activated protein kinase (MAPK) signaling, an effect that is abolished in cells expressing mutant mGluR1 lacking intact caveolin binding motifs. Neurons from caveolin-1 knock-out mice show enhanced basal ERK1/2 phosphorylation and prolonged ERK1/2 activation in response to stimulation with DHPG [(RS)-3,5-dihydroxyphenylglycine], a group I mGluR-selective agonist. Together, these findings underscore the importance of caveolar rafts in neurons and suggest that this pathway might play an important role in synapse formation and plasticity.


Subject(s)
Caveolin 1/physiology , Membrane Microdomains/physiology , Receptors, AMPA/physiology , Signal Transduction/physiology , Animals , Binding Sites/physiology , Caveolin 1/metabolism , Cell Line , Cells, Cultured , Endocytosis/physiology , Humans , Membrane Microdomains/metabolism , Mice , Neuronal Plasticity/physiology , Protein Transport/physiology , Rats , Receptors, AMPA/metabolism , Synapses/physiology
15.
Bioinformation ; 3(6): 275-8, 2009.
Article in English | MEDLINE | ID: mdl-19255648

ABSTRACT

Atrial fibrillation (AF) is a frequent chronic dysrythmia with an incidence that increases with age (>40). Because of its medical and socio-economic impacts it is expected to become an increasing burden on most health care systems. AF is a multi-factorial disease for which the identification of subtypes is warranted. Novel approaches based on the broad concepts of systems biology may overcome the blurred notion of normal and pathological phenotype, which is inherent to high throughput molecular arrays analysis. Here we apply an internal contrast algorithm on AF patient data with an analytical focus on potential entry pathways into the disease. We used a RMA (Robust Multichip Average) normalized Affymetrix micro-array data set from 10 AF patients (geo_accession #GSE2240). Four series of probes were selected based on physiopathogenic links with AF entryways: apoptosis (remodeling), MAP kinase (cell remodeling), OXPHOS (ability to sustain hemodynamic workload) and glycolysis (ischemia). Annotated probe lists were polled with Bioconductor packages in R (version 2.7.1). Genetic profile contrasts were analysed with hierarchical clustering and principal component analysis. The analysis revealed distinct patient groups for all probe sets. A substantial part (54% till 67%) of the variance is explained in the first 2 principal components. Genes in PC1/2 with high discriminatory value were selected and analyzed in detail. We aim for reliable molecular stratification of AF. We show that stratification is possible based on physiologically relevant gene sets. Genes with high contrast value are likely to give pathophysiological insight into permanent AF subtypes.

16.
J Neurochem ; 108(6): 1515-25, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19183249

ABSTRACT

Regulated trafficking of neurotransmitter receptors is critical to normal neurodevelopment and neuronal signaling. Group I mGluRs (mGluR1/5 and their splice variants) are G protein-coupled receptors enriched at excitatory synapses, where they serve to modulate glutamatergic transmission. The mGluR1 splice variants mGluR1a and mGluR1b are broadly expressed in the central nervous system and differ in their signaling and trafficking properties. Several proteins have been identified that selectively interact with mGluR1a and participate in receptor trafficking but no proteins interacting with mGluR1b have thus far been reported. We have used a proteomic strategy to isolate and identify proteins that co-purify with mGluR1b in Madin-Darby Canine Kidney (MDCK) cells, an established model system for trafficking studies. Here, we report the identification of 10 novel candidate mGluR1b-interacting proteins. Several of the identified proteins are structural components of the cell cytoskeleton, while others serve as cytoskeleton-associated adaptors and motors or endoplasmic reticulum-associated chaperones. Findings from this work will help unravel the complex cellular mechanisms underlying mGluR trafficking under physiological and pathological conditions.


Subject(s)
Proteomics/methods , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , Amino Acid Sequence , Animals , Cell Line, Transformed , Dogs , Humans , Immunoprecipitation/methods , Models, Molecular , Protein Binding , Protein Interaction Mapping , Protein Transport , Proteins/chemistry , Proteins/metabolism , Receptors, Metabotropic Glutamate/chemistry , Transfection/methods
17.
Neuron ; 59(1): 1-3, 2008 Jul 10.
Article in English | MEDLINE | ID: mdl-18614022

ABSTRACT

Neuroscientists have been looking for good examples linking neuronal activity to gene expression/regulation involved in synaptic plasticity and the formation of long-term memories. New findings from Park et al. and Waung et al. in this issue of Neuron show that fast dendritic translation of the immediate-early gene Arc/Arg3.1 is involved in hippocampal mGluR-LTD, a protein synthesis-dependent form of plasticity.


Subject(s)
Neuronal Plasticity/physiology , Neurons/metabolism , Protein Biosynthesis/physiology , Animals , Gene Expression Regulation/physiology , Hippocampus/cytology
18.
Europace ; 10(9): 1102-4, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18559333

ABSTRACT

We describe a case of a 50-year-old man with advanced atrioventricular block treated successfully with His-bundle pacing via a persistent left superior vena cava draining into the coronary sinus.


Subject(s)
Atrioventricular Block/therapy , Bundle of His , Cardiac Pacing, Artificial/methods , Vena Cava, Superior/abnormalities , Humans , Male , Middle Aged
19.
J Neurosci ; 24(45): 10138-48, 2004 Nov 10.
Article in English | MEDLINE | ID: mdl-15537884

ABSTRACT

NMDA receptors (NMDARs) colocalize with postsynaptic density protein-95 (PSD-95), a multivalent synaptic scaffolding protein and core component of the postsynaptic density, at excitatory synapses. Although much is known about the identity and properties of scaffolding proteins, little is known about their actions on NMDAR function. Here we show that association of PSD-95 with NMDARs modulates channel gating and surface expression. PSD-95 increases the number of functional channels at the cell surface and channel opening rate of NMDARs, with little or no change in conductance, reversal potential, or mean open time. We show further that PSD-95 increases NMDAR surface expression by increasing the rate of channel insertion and decreasing the rate of channel internalization. The PDZ (PSD-95, discs large, zona occludens-1) binding motif at the distal end of the NR2 C-terminal tail is critical to the actions of PSD-95 on NMDAR function and surface expression. Given that activity bi-directionally modifies synaptic levels of PSD-95, our findings suggest a novel mechanism for activity-dependent regulation of NMDARs at central synapses.


Subject(s)
Ion Channel Gating/physiology , Nerve Tissue Proteins/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Amino Acid Motifs , Animals , Calcium/metabolism , Cell Line , Cell Membrane/metabolism , Cricetinae , Cricetulus , Disks Large Homolog 4 Protein , Dizocilpine Maleate/pharmacology , Female , Guanylate Kinases , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Mice , Mutagenesis, Site-Directed , N-Methylaspartate/pharmacology , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Oocytes , Protein Interaction Mapping , Protein Transport , Rats , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/physiology , Up-Regulation , Xenopus laevis
20.
J Neurosci Res ; 75(4): 472-9, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14743430

ABSTRACT

Metabotropic glutamate receptors (mGluRs) and calcium receptors (CaR) are closely related G protein-coupled receptors (GPCRs). The similar structural and functional properties of mGluRs and CaRs include conserved amino acid residues involved in glutamate binding in mGluRs and Ca2+ binding in the CaR. Furthermore, recent findings have demonstrated that mGluRs can respond to high extracellular Ca2+ (Ca2+(o)) whereas CaR activity is potentiated by L-amino acids. We show that both mGluR1 and mGluR2 are activated by Ca2+(o) in the absence of glutamate in the extracellular media. This activation by Ca2+(o) is antagonized by Mg2+(o). Unlike the CaR, in which the intracellular carboxyl tail has been reported to be involved in Ca2+(o)-dependent activity, the carboxyl tail of mGluRs does not seem to play a role in mediating Ca2+(o) actions. On the other hand, we find that preservation of disulfide bonds in the N-terminal extracellular domain of mGluRs is essential for stimulation by Ca2+(o) as well as glutamate. Because the mGluR1 EC50 for Ca2+(o) is within the physiologic range of Ca2+ in the synaptic cleft, mGluR function is likely regulated by changes in divalent cations caused by synaptic activity under normal or pathologic conditions.


Subject(s)
Calcium/chemistry , Magnesium/chemistry , Receptors, Metabotropic Glutamate/metabolism , Zinc/chemistry , Cations, Divalent/chemistry , Cell Line , Dose-Response Relationship, Drug , Humans , Receptors, Metabotropic Glutamate/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...