Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 218
Filter
1.
Neuropharmacology ; 70: 296-305, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23466331

ABSTRACT

Important functional interactions between the metabotropic glutamate 2 (mGlu2) and 5-hydroxytryptamine2A (5-HT2A) neurotransmitter receptors have been established based on electrophysiological, biochemical and behavioral evidence. Over the last several years, dimerization between 5-HT2A and mGlu2 receptors has been proposed to account for the functional cross-talk between these two receptors in the prefrontal cortex. The pros and cons for the existence of a heteromeric complex between 5-HT2A and mGlu2 receptors will be reviewed here. First, the fundamental criteria needing to establish evidence for heteromeric complexes will be reviewed. Then, the in vitro evidence for and against heteromeric complexes between 5-HT2A and mGlu2 receptors will be discussed in regard to physical and functional interactions. Finally, the data with native in situ mGlu2 and 5-HT2A receptors will be discussed with respect to whether heteromeric complexes or a simple functional interaction between two distinct GPCRs based on brain network activity is the more simple explanation for a range of in vivo data.


Subject(s)
Protein Multimerization/physiology , Receptor, Serotonin, 5-HT2A/metabolism , Receptor, Serotonin, 5-HT2A/physiology , Receptors, Metabotropic Glutamate/metabolism , Receptors, Metabotropic Glutamate/physiology , Animals , Hallucinogens/pharmacology , Humans , Receptor Aggregation/physiology , Receptors, Metabotropic Glutamate/agonists , Schizophrenia/metabolism , Serotonin 5-HT2 Receptor Agonists/pharmacology
2.
FASEB J ; 26(1): 334-45, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21982949

ABSTRACT

The distribution of postsynaptic glutamate receptors has been shown to be regulated by proimmunocytokine tumor necrosis factor α (TNF-α) signaling. The role of TNF-α receptor subtypes in mediating glutamate receptor expression, trafficking, and function still remains unclear. Here, we report that TNF receptor subtypes (TNFR1 and TNFR2) differentially modulate α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR) clustering and function in cultured cortical neurons. We find that genetic deletion of TNFR1 decreases surface expression and synaptic localization of the AMPAR GluA1 subunit, reduces the frequency of miniature excitatory postsynaptic current (mEPSC), and reduces AMPA-induced maximal whole-cell current. In addition, these results are not observed in TNFR2-deleted neurons. The decreased AMPAR expression and function in TNFR1-deleted cells are not significantly restored by short (2 h) or long (24 h) term exposure to TNF-α. In TNFR2-deleted cells, TNF-α promotes AMPAR trafficking to the synapse and increases mEPSC frequency. In the present study, we find no significant change in the GluN1 subunit of NMDAR clusters, location, and mEPSC. This includes applying or withholding the TNF-α treatment in both TNFR1- and TNFR2-deleted neurons. Our results indicate that TNF receptor subtype 1 but not 2 plays a critical role in modulating AMPAR clustering, suggesting that targeting TNFR1 gene might be a novel approach to preventing neuronal AMPAR-mediated excitotoxicity.


Subject(s)
Neurons/physiology , Receptors, AMPA/physiology , Receptors, Tumor Necrosis Factor, Type II/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Synaptic Transmission/physiology , Animals , Brain Ischemia/genetics , Brain Ischemia/metabolism , Brain Ischemia/physiopathology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Disease Models, Animal , Excitatory Postsynaptic Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Patch-Clamp Techniques , Receptor Aggregation/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Signal Transduction/physiology , Synapses/physiology
4.
Neuropsychopharmacology ; 37(2): 338-49, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21814188

ABSTRACT

The capacity of opioids to alleviate inflammatory pain is negatively regulated by the glutamate-binding N-methyl-D-aspartate receptor (NMDAR). Increased activity of this receptor complicates the clinical use of opioids to treat persistent neuropathic pain. Immunohistochemical and ultrastructural studies have demonstrated the coexistence of both receptors within single neurons of the CNS, including those in the mesencephalic periaqueductal gray (PAG), a region that is implicated in the opioid control of nociception. We now report that mu-opioid receptors (MOR) and NMDAR NR1 subunits associate in the postsynaptic structures of PAG neurons. Morphine disrupts this complex by protein kinase-C (PKC)-mediated phosphorylation of the NR1 C1 segment and potentiates the NMDAR-CaMKII, pathway that is implicated in morphine tolerance. Inhibition of PKC, but not PKA or GRK2, restored the MOR-NR1 association and rescued the analgesic effect of morphine as well. The administration of N-methyl-D-aspartic acid separated the MOR-NR1 complex, increased MOR Ser phosphorylation, reduced the association of the MOR with G-proteins, and diminished the antinociceptive capacity of morphine. Inhibition of PKA, but not PKC, CaMKII, or GRK2, blocked these effects and preserved morphine antinociception. Thus, the opposing activities of the MOR and NMDAR in pain control affect their relation within neurons of structures such as the PAG. This finding could be exploited in developing bifunctional drugs that would act exclusively on those NMDARs associated with MORs.


Subject(s)
Pain/physiopathology , Periaqueductal Gray/metabolism , Receptor Aggregation/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Opioid, mu/metabolism , Animals , CHO Cells , Cricetinae , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/physiology , Drug Tolerance/physiology , G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors , G-Protein-Coupled Receptor Kinase 2/physiology , Injections, Intraventricular , Male , Mice , Mice, Inbred ICR , Morphine/administration & dosage , Morphine/antagonists & inhibitors , Morphine/pharmacology , N-Methylaspartate/pharmacology , Neurons/metabolism , Pain/prevention & control , Periaqueductal Gray/drug effects , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/physiology , Receptor Aggregation/drug effects
5.
J Neurosci ; 31(47): 17074-90, 2011 Nov 23.
Article in English | MEDLINE | ID: mdl-22114277

ABSTRACT

Infection by the human immunodeficiency virus (HIV) can result in debilitating neurological syndromes collectively known as HIV-associated neurocognitive disorders. Although the HIV coat protein gp120 has been identified as a potent neurotoxin that enhances NMDA receptor function, the exact mechanisms for this effect are not known. Here we provide evidence that gp120 activates two separate signaling pathways that converge to enhance NMDA-evoked calcium flux by clustering NMDA receptors in modified membrane microdomains. gp120 enlarged and stabilized the structure of lipid microdomains on dendrites by mechanisms that involved a redox-regulated translocation of a sphingomyelin hydrolase (neutral sphingomyelinase-2) to the plasma membrane. A concurrent pathway was activated that accelerated the forward traffic of NMDA receptors by a PKA-dependent phosphorylation of the NR1 C-terminal serine 897 (masks an ER retention signal), followed by a PKC-dependent phosphorylation of serine 896 (important for surface expression). NMDA receptors were preferentially targeted to synapses and clustered in modified membrane microdomains. In these conditions, NMDA receptors were unable to laterally disperse and did not internalize, even in response to strong agonist induction. Focal NMDA-evoked calcium bursts were enhanced by threefold in these regions. Inhibiting membrane modification or NR1 phosphorylation prevented gp120 from accelerating the surface localization of NMDA receptors. Disrupting the structure of membrane microdomains after gp120 treatments restored the ability of NMDA receptors to disperse and internalize. These findings demonstrate that gp120 contributes to synaptic dysfunction in the setting of HIV infection by interfering with NMDA receptor trafficking.


Subject(s)
HIV Envelope Protein gp120/physiology , Membrane Microdomains/metabolism , Receptor Aggregation/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Cells, Cultured , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/virology , Humans , Membrane Microdomains/virology , Protein Transport/physiology , Rats , Rats, Sprague-Dawley
6.
Exp Neurol ; 232(2): 149-53, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21906595

ABSTRACT

Fasciculations are characteristic features of amyotrophic lateral sclerosis (ALS), suggesting abnormally increased excitability of motor axons. Previous nerve excitability studies have shown reduced axonal potassium currents in ALS patients that may contribute to the hyperexcitability and thereby generation of fasciculations. To clarify changes in axonal ion channel expression in motor axons of ALS, we performed immunohistochemistry of potassium and sodium channels in the C7 and L5 ventral/dorsal roots obtained from five autopsy cases of sporadic ALS. Compared to controls, the immunoreactivity of potassium channels (Kv1.2) was markedly reduced in the ventral roots, but normal in the dorsal roots of all the ALS patients. Nodal sodium channel expression was not significantly different in ALS patients and control subjects. Our results show prominently reduced expression of axonal potassium channels, and provide the neuropathological and biological basis for decreased accommodative potassium currents in motor axons of ALS patients. The axonal hyperexcitability would lead to generation of fasciculations, and possibly enhances motor neuron death in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Axons/metabolism , Axons/pathology , Kv1.2 Potassium Channel/metabolism , Aged , Aged, 80 and over , Fasciculation/metabolism , Fasciculation/pathology , Female , Humans , Immunohistochemistry , Male , Middle Aged , Motor Neurons/metabolism , Motor Neurons/pathology , Receptor Aggregation/physiology , Sodium Channels/metabolism , Spinal Nerve Roots/metabolism
7.
J Neurosci ; 31(32): 11547-52, 2011 Aug 10.
Article in English | MEDLINE | ID: mdl-21832185

ABSTRACT

Nestin is expressed in many different progenitors during development including those of the CNS, heart, skeletal muscle, and kidney. The adult expression is mainly restricted to the subependymal zone and dentate gyrus of the brain, the neuromuscular junction, and renal podocytes. In addition, this intermediate filament protein has served as a marker of neural stem/progenitor cells for close to 20 years. Therefore it is surprising that its function in development and adult physiology is still poorly understood. Here we report that nestin deficiency is compatible with normal development of the CNS. The mutant mice, however, show impaired motor coordination. Furthermore, we found that the number of acetylcholine receptor clusters, the nerve length, and the endplate bandwidth are significantly increased in neuromuscular junction area of nestin-deficient mice. This is similar to the phenotype described for deficiency of cyclin-dependent kinase 5 (Cdk5), a candidate downstream affecter of nestin. Moreover, we demonstrate that nestin deficiency can rescue maintenance of acetylcholine receptor clusters in the absence of agrin, similar to Cdk5/agrin double knock-outs, suggesting that the observed nestin deficiency phenotype is the consequence of aberrant Cdk5 activity.


Subject(s)
Central Nervous System/embryology , Central Nervous System/metabolism , Cyclin-Dependent Kinase 5/deficiency , Intermediate Filament Proteins/deficiency , Nerve Tissue Proteins/deficiency , Neuromuscular Junction/metabolism , Receptor Aggregation/physiology , Receptors, Cholinergic/metabolism , Agrin/deficiency , Agrin/genetics , Agrin/metabolism , Animals , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/physiology , Female , Gene Targeting/methods , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/physiology , Male , Mice , Mice, Knockout , Motor Activity/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Nestin , Neuromuscular Junction/physiology , Receptor Aggregation/genetics , Receptors, Cholinergic/genetics , Receptors, Cholinergic/physiology
8.
J Cell Biol ; 191(7): 1381-93, 2010 Dec 27.
Article in English | MEDLINE | ID: mdl-21187331

ABSTRACT

Clathrin-mediated endocytosis (CME) is the major pathway for concentrative uptake of receptors and receptor-ligand complexes (cargo). Although constitutively internalized cargos are known to accumulate into maturing clathrin-coated pits (CCPs), whether and how cargo recruitment affects the initiation and maturation of CCPs is not fully understood. Previous studies have addressed these issues by analyzing the global effects of receptor overexpression on CME or CCP dynamics. Here, we exploit a refined approach using expression of a biotinylated transferrin receptor (bTfnR) and controlling its local clustering using mono- or multivalent streptavidin. We show that local clustering of bTfnR increased CCP initiation. By tracking cargo loading in individual CCPs, we found that bTfnR clustering preceded clathrin assembly and confirmed that bTfnR-containing CCPs mature more efficiently than bTfnR-free CCPs. Although neither the clustering nor the related changes in cargo loading altered the rate of CCP maturation, bTfnR-containing CCPs exhibited significantly longer lifetimes than other CCPs within the same cell. Together these results demonstrate that cargo composition is a key source of the differential dynamics of CCPs.


Subject(s)
Coated Pits, Cell-Membrane/physiology , Receptor Aggregation/physiology , Receptors, Transferrin/metabolism , Adaptor Protein Complex 2/genetics , Adaptor Protein Complex 2/metabolism , Adaptor Protein Complex sigma Subunits/genetics , Adaptor Protein Complex sigma Subunits/metabolism , Animals , Biotin/metabolism , Biotinylation/genetics , Biotinylation/methods , Carbon-Nitrogen Ligases/genetics , Cell Line , Cell Membrane/metabolism , Chlorocebus aethiops , Clathrin Light Chains/genetics , Clathrin Light Chains/metabolism , Coated Pits, Cell-Membrane/ultrastructure , Endocytosis/physiology , Epithelial Cells/cytology , Epithelial Cells/physiology , Escherichia coli Proteins/genetics , Humans , Kinetics , Protein Binding/physiology , Rats , Receptors, Transferrin/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Streptavidin/genetics , Streptavidin/metabolism , Transduction, Genetic
9.
J Immunol ; 185(7): 4179-88, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20817866

ABSTRACT

Free κ L chains (FκLCs) are expressed on the surface of myeloma cells and are being assessed as a therapeutic target for the treatment of multiple myeloma. Despite its clinical potential, the mechanism by which FκLCs interact with membranes remains unresolved. In this study, we show that FκLCs associate with sphingomyelin on the plasma membrane of myeloma cells. Moreover, membrane-bound FκLCs are aggregated, suggesting that aggregation is required for intercalation with membranes. Finally, we propose a model where the binding of FκLCs with sphingomyelin on secretory vesicle membranes is stabilized by self-aggregation, with aggregated FκLCs exposed on the plasma membrane after exocytosis. Although it is well known that protein aggregates bind membranes, this is only the second example of an aggregate being found on the surface of cells that also secrete the protein in its native form. We postulate that many other aggregation-prone proteins may associate with cell membranes by similar mechanisms.


Subject(s)
Immunoglobulin Light Chains/metabolism , Multiple Myeloma/metabolism , Plasma Cells/metabolism , Sphingomyelins/metabolism , Blotting, Western , Cell Line, Tumor , Cell Membrane/chemistry , Cell Membrane/immunology , Cell Membrane/metabolism , Cell Separation , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Multiple Myeloma/pathology , Multiprotein Complexes , Plasma Cells/pathology , Receptor Aggregation/physiology , Transfection
10.
J Neurosci ; 30(37): 12455-65, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20844140

ABSTRACT

At the mammalian skeletal neuromuscular junction, cycling of nicotinic ACh receptors (nAChRs) is critical for the maintenance of a high postsynaptic receptor density. However, the mechanisms that regulate nAChRs recycling in living animals remain unknown. Using in vivo time-lapse imaging, fluorescence recovery after photobleaching, and biochemical pull down assays, we demonstrated that recycling of internalized nAChRs into fully functional and denervated synapses was promoted by both direct muscle stimulation and pharmacologically induced intracellular calcium elevations. Most of internalized nAChRs are recycled directly into synaptic sites. Chelating of intracellular calcium below resting level drastically decreased cycling of nAChRs. Furthermore we found that calcium-dependent AChR recycling is mediated by Ca(2+)/calmodulin-dependent kinase II (CaMKII). Inhibition of CaMKII selectively blocked recycling and caused intracellular accumulation of internalized nAChRs, whereas internalization of surface receptors remained unaffected. Electroporation of CaMKII-GFP isoforms into the sternomastoid muscle showed that muscle-specific CaMKIIßm isoform is highly expressed at the neuromuscular junction (NMJ) and precisely colocalized with nAChRs at crests of synaptic folds while the CaMKIIγ and δ isoforms are poorly expressed in synaptic sites. These results indicate that Ca(2+) along with CaMKII activity are critical for receptor recycling and may provide a mechanism by which the postsynaptic AChR density is maintained at the NMJ in vivo.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Neuromuscular Junction/metabolism , Receptor Aggregation/physiology , Receptors, Cholinergic/metabolism , Animals , Benzylamines/pharmacology , Calcium Signaling/drug effects , Calcium Signaling/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/biosynthesis , Endocytosis/drug effects , Endocytosis/physiology , Female , Fluorescence Recovery After Photobleaching , Isoenzymes/antagonists & inhibitors , Isoenzymes/biosynthesis , Isoenzymes/physiology , Mice , Neuromuscular Junction/cytology , Neuromuscular Junction/enzymology , Protein Kinase Inhibitors/pharmacology , Receptor Aggregation/drug effects , Sulfonamides/pharmacology
11.
Thromb Haemost ; 103(5): 1033-43, 2010 May.
Article in English | MEDLINE | ID: mdl-20216992

ABSTRACT

Incubation at 0 degrees C is known to expose b- N -acetyl-D-glucosamine residues on glycoprotein (GP) Ibalpha inducing receptor clustering and alpha(M)beta(2)-mediated platelet destruction by macrophages. Here we show that incubation at 0/37 degrees C (4 hours at 0 degrees C, followed by 1 hour at 37 degrees C to mimic cold-storage and post-transfusion conditions) triggers a conformational change in the N -terminal flank (NTF, amino acids, aa 1-35) but not in aa 36-282 of GPIbalpha as detected by antibody binding. Addition of the sugar N -acetyl-D-glucosamine (GN) inhibits responses induced by 0/37 degrees C. Incubation at 0 degrees C shifts GPIbalpha from the membrane skeleton to the cytoskeleton. Different GPIbalpha conformations have little effect on VWF/ristocetin-induced aggregation, but arrest of NTF change by GN interferes with agglutination and spreading on a VWF-coated surface under flow. Strikingly, incubation at 0/37 degrees C initiates thromboxane A(2) formation through a von Willebrand factor (VWF)-independent and GPIbalpha-dependent mechanism, as confirmed in VWF- and GPIbalpha-deficient platelets. We conclude that the NTF change induced by 0/37 degrees C incubation reflects clustering of GPIbalpha supports VWF/ristocetin-induced agglutination and spreading and is sufficient to initiate platelet activation in the absence of VWF.


Subject(s)
Acetylglucosamine/metabolism , Blood Platelets/metabolism , Blood Preservation , Platelet Glycoprotein GPIb-IX Complex/metabolism , Platelet Transfusion , von Willebrand Diseases/metabolism , Acetylglucosamine/chemistry , Antibodies, Monoclonal , Blood Platelets/pathology , Cells, Cultured , Cytoskeleton/metabolism , Humans , Macrophage Activation , Macrophage-1 Antigen/metabolism , Platelet Activation , Platelet Glycoprotein GPIb-IX Complex/chemistry , Platelet Glycoprotein GPIb-IX Complex/immunology , Protein Binding , Protein Conformation , Protein Transport , Receptor Aggregation/physiology , Temperature , Thromboxane A2/biosynthesis , Thromboxane A2/genetics , von Willebrand Diseases/blood , von Willebrand Diseases/pathology , von Willebrand Diseases/therapy , von Willebrand Factor/metabolism
12.
J Neurosci ; 30(8): 2935-50, 2010 Feb 24.
Article in English | MEDLINE | ID: mdl-20181591

ABSTRACT

Dopaminergic projections to the striatum, crucial for the correct functioning of this brain region in adulthood, are known to be established early in development, but their role is currently uncharacterized. We demonstrate here that dopamine, by activating D(1)- and/or D(2)-dopamine receptors, decreases the number of functional GABAergic synapses formed between the embryonic precursors of the medium spiny neurons, the principal output neurons of the striatum, with associated changes in spontaneous synaptic activity. Activation of these receptors reduces the size of postsynaptic GABA(A) receptor clusters and their overall cell-surface expression, without affecting the total number of clusters or the size or number of GABAergic nerve terminals. These changes result from an increased internalization of GABA(A) receptors, and are mediated by distinct signaling pathways converging at the level of GABA(A) receptors to cause a transient PP2A/PP1-dependent dephosphorylation. Thus, tonic D(1)- and D(2)-receptor activity limits the extent of collateral inhibitory synaptogenesis between medium spiny neurons, revealing a novel role of dopamine in controlling the development of intrinsic striatal microcircuits.


Subject(s)
Corpus Striatum/embryology , Corpus Striatum/metabolism , Dopamine/metabolism , Neural Inhibition/physiology , Neurogenesis/physiology , Synapses/metabolism , Animals , Cell Differentiation/physiology , Corpus Striatum/cytology , Neural Pathways/cytology , Neural Pathways/embryology , Neural Pathways/metabolism , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Receptor Aggregation/physiology , Receptors, Dopamine/metabolism , Receptors, GABA-A/metabolism , Synaptic Transmission/physiology , Time Factors , gamma-Aminobutyric Acid/metabolism
13.
Neuron ; 65(2): 204-16, 2010 Jan 28.
Article in English | MEDLINE | ID: mdl-20152127

ABSTRACT

The maturation of neuromuscular junctions (NMJs) requires the topological transformation of postsynaptic acetylcholine receptor (AChR)-containing structures from a simple plaque to an elaborate structure composed of pretzel-like branches. This maturation process results in the precise apposition of the presynaptic and postsynaptic specializations. However, little is known about the molecular mechanisms underlying the plaque-to-pretzel transition of AChR clusters. In this study, we identify an essential role for the RhoGEF ephexin1 in the maturation of AChR clusters. Adult ephexin1(-/-) mice exhibit severe muscle weakness and impaired synaptic transmission at the NMJ. Intriguingly, when ephexin1 expression is deficient in vivo, the NMJ fails to mature into the pretzel-like shape, and such abnormalities can be rescued by re-expression of ephexin1. We further demonstrate that ephexin1 regulates the stability of AChR clusters in a RhoA-dependent manner. Taken together, our findings reveal an indispensible role for ephexin1 in regulating the structural maturation and neurotransmission of NMJs.


Subject(s)
Guanine Nucleotide Exchange Factors/physiology , Neuromuscular Junction/growth & development , Synaptic Transmission/physiology , Animals , Cells, Cultured , Guanine Nucleotide Exchange Factors/deficiency , Guanine Nucleotide Exchange Factors/genetics , Mice , Mice, Knockout , Myoblasts/physiology , Myoblasts/ultrastructure , Neuromuscular Junction/genetics , Neuromuscular Junction/ultrastructure , Receptor Aggregation/physiology , Receptors, Cholinergic/biosynthesis , Receptors, Cholinergic/physiology , Rho Guanine Nucleotide Exchange Factors , Synaptic Transmission/genetics
14.
J Mol Neurosci ; 41(3): 347-57, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20143275

ABSTRACT

The effects of low and high concentrations of cocaine have been studied in vitro on the trafficking of plasma membrane A(2A) and D(2) immunoreactivities in previously characterized A(2A)-D(2) CHO cell lines. Receptor double immunofluorescence staining was performed with D(2) and A(2A) antibodies, planar lipid rafts immunolabeling with biotinylated cholera toxin subunit B and membrane invaginations with an anti-caveolin-1 antibody. A computer-assisted image analysis demonstrated a substantial and highly significant rise of membrane-associated D(2) immunoreactivity (IR) after 8 h of exposure to a low concentration of cocaine (150 nM). At this low concentration of cocaine, there was also an increase of membrane associated A(2A) immunoreactivity but smaller and less significant. However, this increase became considerably larger and highly significant at 150 microM at which concentration the rise of D(2) immunoreactivity had begun to disappear. It may be suggested that an allosteric action of cocaine at 150 nM on the D(2) receptors may primarily increase the insertion of D(2) monomers, homomers and also of a subpopulation of A(2A)-D(2) heteromers from the cytoplasm into the plasma membrane due to the conformational change induced by cocaine in the D(2) receptor. The planar lipid rafts and the caveolae are only affected by the higher concentrations of cocaine. It is proposed that changes in D(2) and A(2A)-D(2) trafficking induced by allosteric actions of cocaine at D(2) receptors may contribute to the alterations of D(2) signaling found in cocaine abusers.


Subject(s)
Cocaine/pharmacology , Dopamine Plasma Membrane Transport Proteins/physiology , Membrane Microdomains/drug effects , Receptor Aggregation/drug effects , Receptor, Adenosine A2A/metabolism , Receptors, Dopamine D2/metabolism , Animals , Binding Sites/drug effects , CHO Cells , Cocaine/metabolism , Cricetinae , Cricetulus , Dopamine Uptake Inhibitors/pharmacology , Dose-Response Relationship, Drug , Membrane Microdomains/metabolism , Receptor Aggregation/physiology , Transfection
15.
J Neurosci ; 30(1): 13-23, 2010 Jan 06.
Article in English | MEDLINE | ID: mdl-20053883

ABSTRACT

CollagenQ (ColQ) plays an important structural role at vertebrate neuromuscular junctions (NMJs) by anchoring and accumulating acetylcholinesterase (AChE) in the extracellular matrix (ECM). Moreover, ColQ interacts with perlecan/dystroglycan and the muscle-specific receptor tyrosine kinase (MuSK), key molecules in the NMJ formation. MuSK promotes acetylcholine receptor (AChR) clustering in a process mediated by rapsyn, a cytoplasmic protein that stimulates AChR packing in clusters and regulates synaptic gene transcription. Here, we investigated a regulatory role for ColQ by comparing the clustering and expression of synaptic proteins in wild type and ColQ-deficient muscle cells in culture and at NMJ. We show first that AChR clusters are smaller and more densely packed in the absence of ColQ both in vitro and in vivo. Second, we find that like AChRs and rapsyn, MuSK mRNA levels are increased in cultured cells but not in muscles lacking ColQ. However, membrane-bound MuSK is decreased both in vitro and in vivo suggesting that ColQ controls MuSK sorting or stabilization in the muscle membrane. In line with this, our data show that activation of the MuSK signaling pathway is altered in the absence of ColQ leading to (1) perturbation of AChR clustering and/or beta-AChR subunit phosphorylation and (2) modifications of AChR mRNA level due to the lack of ColQ-MuSK interaction. Together, our results demonstrate that ColQ, in addition to its structural role, has important regulatory functions at the synapse by controlling AChR clustering and synaptic gene expression through its interaction with MuSK.


Subject(s)
Acetylcholinesterase/physiology , Cell Differentiation/physiology , Collagen/physiology , Neuromuscular Junction/physiology , Presynaptic Terminals/physiology , Acetylcholinesterase/chemistry , Acetylcholinesterase/metabolism , Animals , COS Cells , Cell Line , Cells, Cultured , Chlorocebus aethiops , Collagen/chemistry , Collagen/metabolism , Mice , Mice, Knockout , Neuromuscular Junction/cytology , Rats , Receptor Aggregation/physiology , Receptor Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Cholinergic/metabolism , Receptors, Cholinergic/physiology , Synapses/physiology
16.
J Mol Neurosci ; 40(1-2): 40-6, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19680821

ABSTRACT

Acetylcholinesterase (AChE) is well known to process different molecular forms via the distinct interacting partners. Proline-rich membrane anchor (PRiMA)-linked tetrameric globular AChE (G4 AChE) is mainly found in the vertebrate brain; however, recent studies from our laboratory have suggested its existence at neuromuscular junctions (nmjs). Both muscle and motor neuron express AChE at the nmjs. In muscle, the expression of PRiMA-linked AChE is down-regulated during myogenic differentiation and by motor neuron innervation. As compared with muscle, spinal cord possessed higher total AChE activity and contained PRiMA-linked AChE forms. The spinal cord expression of this form increased during development. More importantly, PRiMA-linked G4 AChE identified as aggregates localized at nmjs. These findings suggest that the restricted localization of PRiMA-linked G4 AChE at the nmjs could be contributed by the pre-synaptic motor neuron and/or the post-synaptic muscle fiber.


Subject(s)
Acetylcholinesterase/chemistry , Acetylcholinesterase/metabolism , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Neuromuscular Junction/enzymology , Spinal Cord/enzymology , Animals , Cell Differentiation/genetics , Motor Neurons/enzymology , Motor Neurons/ultrastructure , Muscle, Skeletal/enzymology , Muscle, Skeletal/growth & development , Muscle, Skeletal/innervation , Neuromuscular Junction/growth & development , Neuromuscular Junction/ultrastructure , Presynaptic Terminals/enzymology , Presynaptic Terminals/ultrastructure , Protein Conformation , RNA, Messenger/metabolism , Rats , Receptor Aggregation/physiology , Spinal Cord/growth & development , Spinal Cord/ultrastructure , Synaptic Membranes/enzymology , Synaptic Membranes/ultrastructure , Synaptic Transmission/physiology , Up-Regulation/genetics
17.
Neuroscience ; 163(1): 222-32, 2009 Sep 29.
Article in English | MEDLINE | ID: mdl-19482062

ABSTRACT

At the developing vertebrate neuromuscular junction, the acetylcholine receptor becomes aggregated at high density in the postsynaptic muscle membrane. Receptor localization is regulated by the motoneuron-derived factor, agrin, and requires an intracellular, scaffolding protein called rapsyn. However, it remains unclear where rapsyn binds on the acetylcholine receptor and how their interaction is regulated. In this study, we identified rapsyn's binding site on the acetylcholine receptor using chimeric constructs where the intracellular domain of CD4 was substituted for the major intracellular loop of each mouse acetylcholine receptor subunit. When expressed in heterologous cells, we found that rapsyn clustered and cytoskeletally anchored CD4-alpha, beta and epsilon subunit loops but not CD4-delta loop. Rapsyn-mediated clustering and anchoring was highest for beta loop, followed by epsilon and alpha, suggesting that rapsyn interacts with the loops with different affinities. Moreover, by making deletions within the beta subunit intracellular loop, we show that rapsyn interacts with the alpha-helical region, a secondary structural motif present in the carboxyl terminal portion of the subunit loops. When expressed in muscle cells, rapsyn co-immunoprecipitated together with a CD4-alpha helical region chimera, independent of agrin signaling. Together, these findings demonstrate that rapsyn interacts with the acetylcholine receptor via an alpha-helical structural motif conserved between the alpha, beta and epsilon subunits. Binding at this site likely mediates the critical rapsyn interaction involved in localizing the acetylcholine receptor at the neuromuscular junction.


Subject(s)
Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Neuromuscular Junction/metabolism , Receptors, Cholinergic/metabolism , Synaptic Membranes/metabolism , Agrin/metabolism , Animals , Binding Sites/physiology , COS Cells , Chlorocebus aethiops , Conserved Sequence/physiology , Intracellular Fluid/metabolism , Mice , Muscle, Skeletal/innervation , Muscle, Skeletal/ultrastructure , Mutant Chimeric Proteins/chemistry , Mutant Chimeric Proteins/metabolism , Neuromuscular Junction/ultrastructure , Protein Binding/physiology , Protein Structure, Secondary/physiology , Protein Structure, Tertiary/physiology , Protein Subunits/metabolism , Receptor Aggregation/physiology , Receptors, Cholinergic/chemistry , Signal Transduction/physiology , Synaptic Membranes/ultrastructure
18.
J Biol Chem ; 284(24): 16693-16703, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19386600

ABSTRACT

A functional collaboration between growth factor receptors such as platelet derived growth factor receptor (PDGFR) and integrins is required for effective signal transduction in response to soluble growth factors. However, the mechanisms of synergistic PDGFR/integrin signaling remain poorly understood. Our previous work showed that cell surface tissue transglutaminase (tTG) induces clustering of integrins and amplifies integrin signaling by acting as an integrin binding adhesion co-receptor for fibronectin. Here we report that in fibroblasts tTG enhances PDGFR-integrin association by interacting with PDGFR and bridging the two receptors on the cell surface. The interaction between tTG and PDGFR reduces cellular levels of the receptor by accelerating its turnover. Moreover, the association of PDGFR with tTG causes receptor clustering, increases PDGF binding, promotes adhesion-mediated and growth factor-induced PDGFR activation, and up-regulates downstream signaling. Importantly, tTG is required for efficient PDGF-dependent proliferation and migration of fibroblasts. These results reveal a previously unrecognized role for cell surface tTG in the regulation of the joint PDGFR/integrin signaling and PDGFR-dependent cell responses.


Subject(s)
Fibroblasts/cytology , Fibroblasts/enzymology , Integrins/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Transglutaminases/metabolism , Animals , Cell Adhesion/physiology , Cell Division/physiology , Cell Movement/physiology , Cells, Cultured , Dermis/cytology , Down-Regulation/physiology , Humans , Mice , NIH 3T3 Cells , Receptor Aggregation/physiology , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptors, Cell Surface/metabolism , Signal Transduction/physiology , Transglutaminases/genetics , Up-Regulation/physiology , Wound Healing/physiology
19.
Free Radic Biol Med ; 46(5): 543-54, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19133328

ABSTRACT

Vitamin E is the last of all vitamins whose essentiality is not yet understood. Its widely accepted role as a lipophilic antioxidant has been questioned, since proof of its in vivo relevance remained scarce. The influence of vitamin E on biomarkers of oxidative stress in vivo is inconsistent and metabolites of vitamin E having reacted as an antioxidant are hardly detectable. Novel functions of vitamin E include the regulation of enzymes, most of which are membrane bound or activated by membrane recruitment. Also, expression of genes responds to vitamin E. The search for a transcription factor common to all regulated genes failed so far and a receptor that specifically binds vitamin E has not yet been identified. According to microarray data, pathways preferentially affected by the vitamin E status are the inflammatory response and cellular traffic. A role of vitamin E in cellular trafficking could best explain the neurological symptoms seen in vitamin E deficiency. Emerging knowledge on vitamin E is compiled here with the perspective to unravel the molecular mechanisms that could more likely explain the essentiality of the vitamin than its ability to scavenge free radicals.


Subject(s)
Enzyme Activation/physiology , Neurodegenerative Diseases/metabolism , Receptor Aggregation/physiology , Vitamin E Deficiency/physiopathology , Vitamin E/physiology , Animals , Apoptosis Regulatory Proteins/physiology , Cell Movement/physiology , Female , Gene Expression Regulation, Enzymologic , Humans , Neurodegenerative Diseases/genetics , Placental Circulation/physiology , Polymorphism, Genetic , Pregnancy , Vitamin E Deficiency/genetics , Vitamin E Deficiency/pathology
20.
J Exp Med ; 206(1): 195-207, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-19124655

ABSTRACT

Aggregation of the high affinity IgE receptor (Fc epsilonRI) activates a cascade of signaling events leading to mast cell activation. Subsequently, inhibitory signals are engaged for turning off activating signals. We identified that regulator of calcineurin (Rcan) 1 serves as a negative regulator for turning off Fc epsilonRI-mediated mast cell activation. Fc epsilonRI-induced Rcan1 expression was identified by suppression subtractive hybridization and verified by real-time quantitative polymerase chain reaction and Western blotting. Deficiency of Rcan1 led to increased calcineurin activity, increased nuclear factor of activated T cells and nuclear factor kappaB activation, increased cytokine production, and enhanced immunoglobulin E-mediated late-phase cutaneous reactions. Forced expression of Rcan1 in wild-type or Rcan1-deficient mast cells reduced Fc epsilonRI-mediated cytokine production. Rcan1 deficiency also led to increased Fc epsilonRI-mediated mast cell degranulation and enhanced passive cutaneous anaphylaxis. Analysis of the Rcan1 promoter identified a functional Egr1 binding site. Biochemical and genetic evidence suggested that Egr1 controls Rcan1 expression. Our results identified Rcan1 as a novel inhibitory signal in Fc epsilonRI-induced mast cell activation and established a new link of Egr1 and Rcan1 in Fc epsilonRI signaling.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Mast Cells/physiology , Muscle Proteins/physiology , Receptors, IgE/physiology , Signal Transduction/physiology , Animals , Blotting, Western , Calcium-Binding Proteins , Cell Degranulation/genetics , Cell Degranulation/immunology , Cells, Cultured , Chromatin Immunoprecipitation , Cytokines/genetics , Cytokines/metabolism , Dinitrofluorobenzene/immunology , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Gene Expression , Hypersensitivity/genetics , Hypersensitivity/metabolism , Hypersensitivity/pathology , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Mast Cells/cytology , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Muscle Proteins/deficiency , Muscle Proteins/genetics , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Receptor Aggregation/physiology , Reverse Transcriptase Polymerase Chain Reaction , Serum Albumin, Bovine/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...