Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 756: 273-81, 2011.
Article in English | MEDLINE | ID: mdl-21870232

ABSTRACT

It is now well understood that G protein-coupled receptor (GPCR)-mediated cell signalling is subject to extensive spatial-temporal control, and that a meaningful understanding of this complexity requires techniques to study signalling at the molecular and sub-cellular level. This complexity in cell signal pattern begins with ligand binding to the receptor and its coupling to a variety of different effector systems. These signal transduction cascades within a cell involve a very complex series of molecular events requiring the generation of multiple second messenger responses and the activation a multiple effector proteins. In the present chapter, we will describe methodology for the simultaneous assessment of the spatial-temporal measurement of increases in intracellular Ca2+ concentrations and the activation of protein kinase C (PKC) in response to the agonist activation of a Gαq/11-coupled GPCR. Specifically, we will describe a confocal imaging approach to simultaneously measure oscillilations in intracellular Ca2+ levels and PKC translocation to the plasma membrane in response to mGluR1 stimulation in transiently transfected human embryonic kidney (HEK293) cells. The changes in intracellular Ca2+ were imaged using the fluorescent indicator Oregon Green 488 BAPTA and a recombinant PKCßII-DsRed fusion protein was used to image the sub-cellular distribution of the PKCßII isoform.


Subject(s)
Calcium Signaling , Calcium/analysis , Calcium/metabolism , Microscopy, Confocal/methods , Protein Kinase C/analysis , Protein Kinase C/metabolism , Carboxylic Acids/analysis , Cell Line , Humans , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Protein Kinase C/genetics , Protein Kinase C beta , Protein Transport , Receptors, G-Protein-Coupled/metabolism , Receptors, Metabotropic Glutamate/metabolism , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection
2.
Mol Pharmacol ; 79(1): 175-84, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20943774

ABSTRACT

The human angiotensin II type 1 receptor (AT1R) is a member of the G protein-coupled receptor (GPCR) superfamily and represents an important target for cardiovascular therapeutic intervention. Agonist-activation of the AT1R induces ß-arrestin-dependent endocytosis to early endosomes in which the receptor resides as a protein complex with the Rab GTPase Rab5. In the present study, we examined whether other Rab GTPases that regulate receptor trafficking through endosomal compartments also bind to the AT1R. We find that Rab4, Rab7, and Rab11 all bind to the last 10 amino acid residues of the AT1R carboxyl-terminal tail. Rab11 binds AT1R more effectively than Rab5, whereas Rab4 binds less effectively than Rab5. Alanine scanning mutagenesis reveals that proline 354 and cysteine 355 contribute to Rab protein binding, and mutation of these residues does not affect G protein coupling. We find that the Rab GTPases each compete with one another for receptor binding and that although Rab4 interacts poorly with the AT1R, it effectively displaces Rab11 from the receptor. In contrast, Rab11 overexpression does not prevent Rab4 binding to the AT1R. Overexpression of wild-type Rab4, but not Rab11, facilitates AT1R dephosphorylation, and a constitutively active Rab4-Q67L mutant reduces AT1R desensitization and promotes AT1R resensitization. Taken together, our data indicate that multiple Rab GTPases bind to a motif localized to the distal end of the AT1R tail and that increased Rab4 activity may contribute to the regulation AT1R desensitization and dephosphorylation.


Subject(s)
Receptor, Angiotensin, Type 1/metabolism , rab GTP-Binding Proteins/metabolism , rab4 GTP-Binding Proteins/physiology , Binding Sites/physiology , HEK293 Cells , Humans , Phosphorylation/physiology , Protein Binding/physiology , Receptor, Angiotensin, Type 1/chemistry , rab GTP-Binding Proteins/chemistry , rab4 GTP-Binding Proteins/chemistry
3.
Nat Neurosci ; 13(5): 622-9, 2010 May.
Article in English | MEDLINE | ID: mdl-20383137

ABSTRACT

Stress and anxiety disorders are risk factors for depression and these behaviors are modulated by corticotrophin-releasing factor receptor 1 (CRFR1) and serotonin receptor (5-HT(2)R). However, the potential behavioral and cellular interaction between these two receptors is unclear. We found that pre-administration of corticotrophin-releasing factor (CRF) into the prefrontal cortex of mice enhanced 5-HT(2)R-mediated anxiety behaviors in response to 2,5-dimethoxy-4-iodoamphetamine. In both heterologous cell cultures and mouse cortical neurons, activation of CRFR1 also enhanced 5-HT(2) receptor-mediated inositol phosphate formation. CRFR1-mediated increases in 5-HT(2)R signaling were dependent on receptor internalization and receptor recycling via rapid recycling endosomes, resulting in increased expression of 5-HT(2)R on the cell surface. Sensitization of 5-HT(2)R signaling by CRFR1 required intact PDZ domain-binding motifs at the end of the C-terminal tails of both receptor types. These data suggest a mechanism by which CRF, a peptide known to be released by stress, enhances anxiety-related behavior via sensitization of 5-HT(2)R signaling.


Subject(s)
Anxiety/metabolism , Receptors, Corticotropin-Releasing Hormone/physiology , Receptors, Serotonin, 5-HT2/metabolism , Signal Transduction/physiology , Amphetamines/pharmacology , Animals , Anxiety/drug therapy , Anxiety/physiopathology , Behavior, Animal/drug effects , Behavior, Animal/physiology , Biotinylation/methods , Cells, Cultured , Corticotropin-Releasing Hormone/pharmacology , Cyclic AMP/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Embryo, Mammalian , Fluorobenzenes/pharmacology , Hormones/pharmacology , Humans , Inositol Phosphates/metabolism , Ionophores/pharmacology , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Monensin/pharmacology , Mutation/genetics , Neurons , Piperidines/pharmacology , Prefrontal Cortex/cytology , Rats , Reaction Time/drug effects , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Serotonin, 5-HT2/genetics , Serotonin/pharmacology , Serotonin 5-HT2 Receptor Agonists , Serotonin 5-HT2 Receptor Antagonists , Serotonin Agents/pharmacology , Signal Transduction/drug effects , Transfection
4.
Mol Brain ; 3: 4, 2010 Jan 21.
Article in English | MEDLINE | ID: mdl-20180987

ABSTRACT

Group I metabotropic glutamate receptors (mGluRs) are coupled via Galphaq/11 to the activation of phospholipase Cbeta, which hydrolyzes membrane phospholipids to form inositol 1,4,5 trisphosphate and diacylglycerol. This results in the release of Ca2+ from intracellular stores and the activation of protein kinase C. The activation of Group I mGluRs also results in ERK1/2 phosphorylation. We show here, that the proline-rich tyrosine kinase 2 (Pyk2) interacts with both mGluR1 and mGluR5 and is precipitated with both receptors from rat brain. Pyk2 also interacts with GST-fusion proteins corresponding to the second intracellular loop and the distal carboxyl-terminal tail domains of mGluR1a. Pyk2 colocalizes with mGluR1a at the plasma membrane in human embryonic kidney (HEK293) cells and with endogenous mGluR5 in cortical neurons. Pyk2 overexpression in HEK293 results in attenuated basal and agonist-stimulated inositol phosphate formation in mGluR1 expressing cells and involves a mechanism whereby Pyk2 displaces Galphaq/11 from the receptor. The activation of endogenous mGluR1 in primary mouse cortical neuron stimulates ERK1/2 phosphorylation. Treatments that prevent Pyk2 phosphorylation in cortical neurons, and the overexpression of Pyk2 dominant-negative and catalytically inactive Pyk2 mutants in HEK293 cells, prevent ERK1/2 phosphorylation. The Pyk2 mediated activation of ERK1/2 phosphorylation is also Src-, calmodulin- and protein kinase C-dependent. Our data reveal that Pyk2 couples the activation mGluRs to the mitogen-activated protein kinase pathway even though it attenuates mGluR1-dependent G protein signaling.


Subject(s)
Focal Adhesion Kinase 2/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction/physiology , Animals , Enzyme Activation , Enzyme Inhibitors/metabolism , Excitatory Amino Acid Agonists/metabolism , Focal Adhesion Kinase 2/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , HEK293 Cells , Humans , Imidazoles/metabolism , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Neurons/metabolism , Phosphorylation , Protein Conformation , Quisqualic Acid/metabolism , RNA, Small Interfering/metabolism , Rats , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/chemistry , Receptors, Metabotropic Glutamate/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tyrphostins/metabolism
5.
Mol Pharmacol ; 77(3): 388-95, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20018811

ABSTRACT

The angiotensin II type 1 receptor (AT(1)R) plays an important role in cardiovascular function and as such represents a primary target for therapeutic intervention. The AT(1)R has traditionally been considered to be coupled to the activation of phospholipase C (PLC) beta via its association with G alpha(q/11), leading to increases in intracellular inositol phosphate (IP) and release of calcium from intracellular stores. In the present study, we investigated whether the small GTPase RalA contributed to the regulation of AT(1)R endocytosis and signaling. We find that neither RalA nor RalB is required for the endocytosis of the AT(1)R, but that RalA expression is required for AT(1)R-stimulated IP formation but not 5-HT(2A) receptor-mediated IP formation. AT(1)R-activated IP formation is lost in the absence of Ral guanine nucleotide dissociation stimulator (RalGDS), and requires the beta-arrestin-dependent plasma membrane translocation of RalGDS. G alpha(q/11) small interfering RNA (siRNA) treatment also significantly attenuates both AT(1)R- and 5-HT(2A) receptor-stimulated IP formation after 30 min of agonist stimulation. PLC-delta1 has been reported to be activated by RalA, and we show that AT(1)R-stimulated IP formation is attenuated after PLC-delta 1 siRNA treatment. Taken together, our results provide evidence for a G protein-coupled recepto-activated and RalGDS/Ral-mediated mechanism for PLC-delta 1 stimulation.


Subject(s)
Phospholipase C delta/metabolism , Receptor, Angiotensin, Type 1/metabolism , ral GTP-Binding Proteins/metabolism , Cell Line , Enzyme Activation/physiology , Humans , Monomeric GTP-Binding Proteins/analysis , Monomeric GTP-Binding Proteins/metabolism , Phospholipase C delta/analysis , Protein Binding/physiology , Receptor, Angiotensin, Type 1/analysis , ral GTP-Binding Proteins/analysis
6.
J Biol Chem ; 284(42): 28986-94, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19717561

ABSTRACT

Group I metabotropic glutamate receptors (mGluRs) are coupled via phospholipase Cbeta to the hydrolysis of phosphoinositides and function to modulate neuronal excitability and synaptic transmission at glutamatergic synapses. The desensitization of Group I mGluR signaling is thought to be mediated primarily via second messenger-dependent protein kinases and G protein-coupled receptor kinases. We show here that both mGluR1 and mGluR5 interact with the calcineurin inhibitor protein (CAIN). CAIN is co-immunoprecipitated in a complex with Group I mGluRs from both HEK 293 cells and mouse cortical brain lysates. Purified CAIN and its C-terminal domain specifically interact with glutathione S-transferase fusion proteins corresponding to the second intracellular loop and the distal C-terminal tail domains of mGluR1. The interaction of CAIN with mGluR1 could also be blocked using a Tat-tagged peptide corresponding to the mGluR1 second intracellular loop domain. Overexpression of full-length CAIN attenuates the agonist-stimulated endocytosis of both mGluR1a and mGluR5a in HEK 293 cells, but expression of the CAIN C-terminal domain does not alter mGluR5a internalization. In contrast, overexpression of either full-length CAIN or the CAIN C-terminal domain impairs agonist-stimulated inositol phosphate formation in HEK 293 cells expressing mGluR1a. This CAIN-mediated antagonism of mGluR1a signaling appears to involve the disruption of receptor-Galpha(q/11) complexes. Taken together, these observations suggest that the association of CAIN with intracellular domains involved in mGluR/G protein coupling provides an additional mechanism by which Group I mGluR endocytosis and signaling are regulated.


Subject(s)
Calcineurin/physiology , Receptors, Metabotropic Glutamate/metabolism , Adaptor Proteins, Signal Transducing , Calcineurin/chemistry , Calcium/metabolism , Cell Line , Endocytosis , Humans , Inositol Phosphates/chemistry , Microscopy, Fluorescence/methods , Models, Biological , Phosphorylation , Plasmids/metabolism , Protein Binding , Protein Kinase C/metabolism , Protein Structure, Tertiary , Signal Transduction
7.
J Neurosci ; 27(47): 12989-99, 2007 Nov 21.
Article in English | MEDLINE | ID: mdl-18032672

ABSTRACT

Oxidative stress has been implicated as a key trigger of neuronal apoptosis in stroke and neurodegenerative conditions such as Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis. The Bcl-2 homology 3 (BH3)-only subfamily of Bcl-2 genes consists of multiple members that can be activated in a cell-type- and stimulus-specific manner to promote cell death. In the present study, we demonstrate that, in cortical neurons, oxidative stress induces the expression of the BH3-only members Bim, Noxa, and Puma. Importantly, we have determined that Puma-/- neurons, but not Bim-/- or Noxa-/- neurons, are remarkably resistant to the induction of apoptosis by multiple oxidative stressors. Furthermore, we have determined that Bcl-2-associated X protein (Bax) is also required for oxidative stress induced cell death and that Puma plays a dominant role in regulating Bax activation. Specifically, we have established that the induction of Puma, but not Bim or Noxa, is necessary and sufficient to induce a conformational change in Bax to its active state, its translocation to the mitochondria and mitochondrial membrane permeabilization. Finally, we demonstrate that whereas both Puma and Bim(EL) can bind to the antiapoptotic family member Bcl-X(L), only Puma was found to associate with Bax. This suggests that in addition to neutralizing antiapoptotic members, Puma may play a dominant role by complexing with Bax and directly promoting its activation. Overall, we have identified Puma as a dominant regulator of oxidative stress induced Bax activation and neuronal apoptosis, and suggest that Puma may be an effective therapeutic target for the treatment of a number of neurodegenerative conditions.


Subject(s)
Apoptosis/physiology , Neurons/metabolism , Oxidative Stress/physiology , Tumor Suppressor Proteins/physiology , bcl-2-Associated X Protein/metabolism , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins , Cells, Cultured , Cerebral Cortex/chemistry , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Gene Expression Regulation/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/chemistry , Neurons/pathology , Oxidative Stress/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/physiology
8.
J Biol Chem ; 281(36): 26340-9, 2006 Sep 08.
Article in English | MEDLINE | ID: mdl-16831865

ABSTRACT

The angiotensin II type 1A receptor (AT(1A)R) plays an important role in cardiovascular function and as such represents a primary target for therapeutic intervention. The AT(1A)R is coupled via G(q) to the activation of phospholipase C, the hydrolysis of phosphoinositides, release of calcium from intracellular stores, and the activation of protein kinase C (PKC). We show here that PKCbetaI and PKCbetaII exhibit different membrane translocation patterns in response to AT(1A)R agonist activation. Whereas PKCbetaII translocation to the membrane is transient, PKCbetaI displays additional translocation responses: persistent membrane localization and oscillations between the membrane and cytosol following agonist removal. The initial translocation of PKCbetaI requires the release of calcium from intracellular stores and the activation of phospholipase C, but persistent membrane localization is dependent upon extracellular calcium influx. The mutation of any of the three PKC phosphorylation consensus sites (Ser-331, Ser-338, and Ser-348) localized within the AT(1A)R C-tail significantly increases the probability that persistent increases in diacylglycerol levels and PKCbetaI translocation responses will be observed. The persistent increase in AT(1A)R-mediated diacylglycerol formation is mediated by the activation of phospholipase D. Although the persistent PKCbetaI membrane translocation response is absolutely dependent upon the PKC activity-dependent recruitment of an extracellular calcium current, it does not require the activation of phospholipase D. Taken together, we show that the patterning of AT(1A)R second messenger response patterns is regulated by heterologous desensitization and PKC isoform substrate specificity.


Subject(s)
Calcium/metabolism , Isoenzymes/metabolism , Phospholipase D/metabolism , Protein Kinase C/metabolism , Receptor, Angiotensin, Type 1/metabolism , Alternative Splicing , Animals , Cell Line , Cell Membrane/metabolism , Diglycerides/metabolism , Enzyme Activation , Humans , Isoenzymes/genetics , Patch-Clamp Techniques , Phospholipase D/genetics , Phosphorylation , Protein Kinase C/genetics , Protein Kinase C beta , Protein Kinase C-delta/metabolism , Protein Transport , RNA, Small Interfering/metabolism , Receptor, Angiotensin, Type 1/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
9.
J Neurochem ; 96(4): 934-49, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16412099

ABSTRACT

The corticotropin releasing factor (CRF) type 1alpha receptor, a member of the G protein-coupled receptor (GPCR) subfamily B, is involved in the aetiology of anxiety and depressive disorders. In the present study, we examined the internalization and trafficking of the CRF1alpha receptor in both human embryonic kidney (HEK)293 cells and primary cortical neurons. We found that CRF1alpha receptor activation leads to the selective recruitment of beta-arrestin2 in both HEK293 cells and neurons. We observed distinct distribution patterns of CRF1alpha receptor and beta-arrestin2 in HEK293 cells and cortical neurons. In HEK293 cells, beta-arrestin2-green fluorescent protein (GFP) co-localized with CRF1alpha receptor in vesicles at the plasma membrane but was dissociated from the receptor in endosomes. In contrast, in primary cortical neurons, beta-arrestin2 and CRF1alpha receptor were internalized in distinct endocytic vesicles. By bioluminescence resonance energy transfer, we demonstrated that beta-arrestin2 association with CRF1alpha receptor was increased in cells transfected with G protein-coupled receptor kinase (GRK)3 and GRK6 and decreased in cells transfected with GRK2 and GRK5. In both HEK293 cells and cortical neurons, internalized CRF1alpha receptor transited from Rab5-positive early endosomes to Rab4-positive recycling endosomes and was not targeted to lysosomes. However, CRF1alpha receptor resensitization was blocked by the overexpression of wild-type, but not dominant-negative, Rab5 and Rab4 GTPases. Taken together, our results suggest that beta-arrestin trafficking differs between HEK293 cells and neurons, and that CRF1alpha receptor resensitization is regulated in an atypical manner by Rab GTPases.


Subject(s)
Arrestins/physiology , Receptors, Corticotropin-Releasing Hormone/physiology , rab GTP-Binding Proteins/metabolism , Adenylyl Cyclases/metabolism , Animals , Cell Line , Cerebral Cortex/physiology , Endocytosis , Green Fluorescent Proteins/metabolism , Humans , Kidney , Kinetics , Luminescence , Mice , Neurons/cytology , Neurons/physiology , Receptors, Corticotropin-Releasing Hormone/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , Transfection , beta-Arrestins
10.
J Biol Chem ; 280(41): 34840-8, 2005 Oct 14.
Article in English | MEDLINE | ID: mdl-16091361

ABSTRACT

Huntington disease is caused by a polyglutamine expansion in the huntingtin protein (Htt) and is associated with excitotoxic death of striatal neurons. Group I metabotropic glutamate receptors (mGluRs) that are coupled to inositol 1,4,5-triphosphate formation and the release of intracellular Ca(2+) stores play an important role in regulating neuronal function. We show here that mGluRs interact with the Htt-binding protein optineurin that is also linked to normal pressure open angled glaucoma and, when expressed in HEK 293 cells, optineurin functions to antagonize agonist-stimulated mGluR1a signaling. We find that Htt is co-precipitated with mGluR1a and that mutant Htt functions to facilitate optineurin-mediated attenuation of mGluR1a signaling. In striatal cell lines derived from Htt(Q111/Q111) mutant knock-in mice mGluR5-stimulated inositol phosphate formation is also severely impaired when compared with striatal cells derived from Htt(Q7/Q7) knock-in mice. In addition, we show that a missense single nucleotide polymorphism optineurin H486R variant previously identified to be associated with glaucoma is selectively impaired in mutant Htt binding. Although optineurin H486R retains the capacity to bind to mGluR1a, optineurin H486R-dependent attenuation of mGluR1a signaling is not enhanced by the expression of mutant Htt. Because G protein-coupled receptor kinase 2 (GRK2) protein expression is relatively low in striatal tissue, we propose that optineurin may substitute for GRK2 in the striatum to mediate mGluR desensitization. Taken together, these studies identify a novel mechanism for mGluR desensitization and an additional biochemical link between altered glutamate receptor signaling and Huntington disease.


Subject(s)
Nerve Tissue Proteins/chemistry , Nuclear Proteins/chemistry , Receptors, Metabotropic Glutamate/metabolism , Transcription Factor TFIIIA/chemistry , Animals , Brain/metabolism , COS Cells , Calcium/metabolism , Cell Cycle Proteins , Cell Line , Cell Survival , Chlorocebus aethiops , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , G-Protein-Coupled Receptor Kinase 2 , Gene Library , Genes, Reporter , Green Fluorescent Proteins/metabolism , Histidine/chemistry , Humans , Huntingtin Protein , Immunoblotting , Immunoprecipitation , Inositol 1,4,5-Trisphosphate/metabolism , Inositol Phosphates/chemistry , Lipids/chemistry , Membrane Transport Proteins , Mice , Mice, Transgenic , Microscopy, Fluorescence , Mutation , Mutation, Missense , Neurons/metabolism , Plasmids/metabolism , Polymorphism, Single Nucleotide , Protein Binding , Rats , Signal Transduction , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Transfection , Two-Hybrid System Techniques , beta-Adrenergic Receptor Kinases/metabolism
11.
J Neurosci ; 24(40): 8752-61, 2004 Oct 06.
Article in English | MEDLINE | ID: mdl-15470141

ABSTRACT

G-protein-coupled receptors play a central role in the regulation of neuronal cell communication. Class 1 metabotropic glutamate receptors (mGluRs) mGluR1a and mGluR5a, which are coupled with the hydrolysis of phosphoinositides, are essential for modulating excitatory neurotransmission at glutamatergic synapses. These receptors are constitutively internalized in heterologous cell cultures, neuronal cultures, and intact neuronal tissues. We show here that the small GTP-binding protein Ral, its guanine nucleotide exchange factor RalGDS (Ral GDP dissociation stimulator), and phospholipase D2 (PLD2) are constitutively associated with class 1 mGluRs and regulate constitutive mGluR endocytosis. Moreover, both Ral and PLD2 are colocalized with mGluRs in endocytic vesicles in both human embryonic kidney 293 (HEK 293) cells and neurons. Ral and PLD2 activity is required for the internalization of class 1 mGluRs but is not required for the internalization of the beta2-adrenergic receptor. Constitutive class 1 mGluR internalization is not dependent on the downstream Ral effector proteins Ral-binding protein 1 and PLD1 or either ADP-ribosylation factors ARF1 or ARF6. The treatment of HEK 293 cells and neurons with small interfering RNA both downregulates PLD2 expression and blocks mGluR1a and mGluR5a endocytosis. The constitutive internalization of mGluR1a and mGluR5a is also attenuated by the treatment of cells with 1-butanol to prevent PLD2-mediated phosphatidic acid formation. We propose that the formation of a mGluR-scaffolded RalGDS/Ral/PLD2 protein complex provides a novel alternative mechanism to beta-arrestins for the constitutive endocytosis of class 1 mGluRs.


Subject(s)
Endocytosis , Neurons/metabolism , Phospholipase D/physiology , Receptors, Metabotropic Glutamate/metabolism , ral GTP-Binding Proteins/physiology , Animals , Cell Line , Cells, Cultured , Humans , Neurons/chemistry , Rats , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/analysis , Signal Transduction , ral GTP-Binding Proteins/analysis
12.
J Biol Chem ; 279(16): 16614-20, 2004 Apr 16.
Article in English | MEDLINE | ID: mdl-14764583

ABSTRACT

Heterotrimeric guanine nucleotide-binding (G) protein-coupled receptor kinases (GRKs) are cytosolic proteins that contribute to the adaptation of G protein-coupled receptor signaling. The canonical model for GRK-dependent receptor desensitization involves GRK-mediated receptor phosphorylation to promote the binding of arrestin proteins that sterically block receptor coupling to G proteins. However, GRK-mediated desensitization, in the absence of phosphorylation and arrestin binding, has been reported for metabotropic glutamate receptor 1 (mGluR1) and gamma-aminobutyric acid B receptors. Here we show that GRK2 mutants impaired in Galphaq/11 binding (R106A, D110A, and M114A), bind effectively to mGluR1a, but do not mediate mGluR1a adaptation. Galphaq/11 is immunoprecipitated as a complex with mGluR1a in the absence of agonist, and either agonist treatment or GRK2 overexpression promotes the dissociation of the receptor/Galphaq/11 complex. However, these mGluR1a/Galphaq/11 interactions are not antagonized by the overexpression of either GRK2 mutants defective in Galphaq/11 binding or RGS4. We have also identified a GRK2-D527A mutant that binds Galphaq/11 in an AlF4(-)-dependent manner but is unable to either bind mGluR1a or attenuate mGluR1a signaling. We conclude that the mechanism underlying GRK2 phosphorylation-independent attenuation of mGluR1a signaling is RH domain-dependent, requiring the binding of GRK2 to both Galphaq/11 and mGluR1a. This serves to coordinate GRK2 interactions with Galphaq/11 and to disrupt receptor/Galphaq/11 complexes. Our findings indicate that GRK2 regulates receptor/G protein interactions, in addition to its traditional role as a receptor kinase.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , GTP-Binding Proteins/metabolism , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction , Binding Sites , Cell Line , Cyclic AMP-Dependent Protein Kinases/genetics , Humans , Models, Molecular , Mutation , Phosphorylation , Receptors, Metabotropic Glutamate/genetics , beta-Adrenergic Receptor Kinases
13.
J Biol Chem ; 279(13): 13110-8, 2004 Mar 26.
Article in English | MEDLINE | ID: mdl-14711821

ABSTRACT

Previous studies have demonstrated that the interaction of the angiotensin II type 1A receptor (AT(1A)R) carboxyl-terminal tail with Rab5a may modulate Rab5a activity, leading to the homotypic fusion of endocytic vesicles. Therefore, we have investigated whether AT(1A)R/Rab5a interactions mediate the retention of AT(1A)R.beta-arrestin complexes in early endosomes and whether the overexpression of Rab7 and Rab11 GTPases influences AT(1A)R lysosomal degradation and plasma membrane recycling. We found that internalized AT(1A)R was retained in Rab5a-positive early endosomes and was neither targeted to lysosomes nor recycled back to the cell surface, whereas a mutant defective in Rab5a binding, AT(1A)R-(1-349), was targeted to lysosomes for degradation. However, the loss of Rab5a binding to the AT(1A)R carboxyl-terminal tail did not promote AT(1A)R recycling. Rather, it was the stable binding of beta-arrestin to the AT(1A)R that prevented, at least in part, AT(1A)R recycling. The overexpression of wild-type Rab7 and Rab7-Q67L resulted in both increased AT(1A)R degradation and AT(1A)R targeting to lysosomes. The Rab7 expression-dependent transition of "putative" AT(1A)R.beta-arrestin complexes to late endosomes was blocked by the expression of dominant-negative Rab5a-S34N. Rab11 overexpression established AT(1A)R recycling and promoted the redistribution of AT(1A)R.beta-arrestin complexes from early to recycling endosomes. Taken together, our data suggest that Rab5, Rab7, and Rab11 work in concert with one another to regulate the intracellular trafficking patterns of the AT(1A)R.


Subject(s)
Receptor, Angiotensin, Type 1/chemistry , rab GTP-Binding Proteins/chemistry , rab5 GTP-Binding Proteins/chemistry , Cell Line , DNA/chemistry , Endocytosis , Endosomes , Epitopes/chemistry , Green Fluorescent Proteins , Humans , Immunoblotting , Luminescent Proteins/metabolism , Lysosomes/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Mutation , Protein Binding , Protein Structure, Tertiary , Time Factors , rab GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
14.
J Biol Chem ; 278(7): 5419-26, 2003 Feb 14.
Article in English | MEDLINE | ID: mdl-12454004

ABSTRACT

Metabotropic glutamate receptors (mGluRs) coupled via Gq to the hydrolysis of phosphoinositides stimulate Ca(2+) and PKCbetaII oscillations in both excitable and non-excitable cells. In the present study, we show that mGluR1a activation stimulates the repetitive plasma membrane translocation of each of the conventional and novel, but not atypical, PKC isozymes. However, despite similarities in sequence and cofactor regulation by diacyglycerol and Ca(2+), conventional PKCs exhibit isoform-specific oscillation patterns. PKCalpha and PKCbetaI display three distinct patterns of activity: (1) agonist-independent oscillations, (2) agonist-stimulated oscillations, and (3) persistent plasma membrane localization in response to mGluR1a activation. In contrast, only agonist-stimulated PKCbetaII translocation responses are observed in mGluR1a-expressing cells. PKCbetaI expression also promotes persistent increases in intracellular diacyglycerol concentrations in response to mGluR1a stimulation without affecting PKCbetaII oscillation patterns in the same cell. PKCbetaII isoform-specific translocation patterns are regulated by specific amino acid residues localized within the C-terminal PKC V5 domain. Specifically, Asn-625 and Lys-668 localized within the V5 domain of PKCbetaII cooperatively suppress PKCbetaI-like response patterns for PKCbetaII. Thus, redundancy in PKC isoform expression and differential decoding of second messenger response provides a novel mechanism for generating cell type-specific responses to the same signal.


Subject(s)
Protein Kinase C/metabolism , Receptors, Metabotropic Glutamate/metabolism , Second Messenger Systems , Cell Line , Humans , Isoenzymes/metabolism , Organ Specificity , Signal Transduction
15.
Neurochem Int ; 41(5): 319-26, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12176073

ABSTRACT

Metabotropic glutamate receptors (mGluRs) constitute an unique subclass of G protein-coupled receptors (GPCRs). These receptors are activated by the excitatory amino acid glutamate and play an essential role in regulating neural development and plasticity. In the present review, we overview the current understanding regarding the molecular mechanisms involved in the desensitization and endocytosis of Group 1 mGluRs as well as the relative contribution of desensitization to the spatial-temporal patterning of glutamate receptor signaling. Similar to what has been reported previously for prototypic GPCRs, mGluRs desensitization is mediated by second messenger-dependent protein kinases and GPCR kinases (GRKs). However, it remains to be determined whether mGluRs phosphorylation by GRKs and beta-arrestin binding are absolutely required for desensitization. Group 1 mGluRs endocytosis is both agonist-dependent and -independent. Agonist-dependent mGluRs internalization is mediated by a beta-arrestin- and dynamin-dependent clathrin-coated vesicle dependent endocytic pathway. The activation of Group 1 mGluRs also results in oscillatory Gq protein-coupling leading to the cyclical activation of phospholipase Cbeta thereby stimulating oscillations in both inositol 1,4,5-triphosphate formation and Ca(2+) release from intracellular stores. These glutamate receptor-stimulated Ca(2+) oscillations are translated into the synchronous activation of protein kinase C (PKC), which has led to the hypothesis that oscillatory mGluRs signaling involves the repetitive phosphorylation of mGluRs by PKC. However, recent experimental evidence suggests that oscillatory signaling is an intrinsic glutamate receptor property that is independent of feedback receptor phosphorylation by PKC. The challenge in the future will be to determine the structural determinants underlying mGluRs-mediated spatial-temporal signaling as well as to understand how complex signaling patterns can be interpreted by cells in both the developing and adult nervous systems.


Subject(s)
Enzyme Activation/physiology , Receptors, Metabotropic Glutamate/physiology , Animals , Arrestins/physiology , Endocytosis/physiology , Humans , Phosphorylation , Receptors, Metabotropic Glutamate/metabolism , Second Messenger Systems/physiology
16.
J Biol Chem ; 277(28): 25266-72, 2002 Jul 12.
Article in English | MEDLINE | ID: mdl-12101219

ABSTRACT

The accepted paradigm for G protein-coupled receptor kinase (GRK)-mediated desensitization of G protein-coupled receptors involves GRK-mediated receptor phosphorylation followed by the binding of arrestin proteins. Although GRKs contribute to metabotropic glutamate receptor 1 (mGluR1) inactivation, beta-arrestins do not appear to be required for mGluR1 G protein uncoupling. Therefore, we investigated whether the phosphorylation of serine and threonine residues localized within the C terminus of mGluR1a is sufficient to allow GRK2-mediated attenuation of mGluR1a signaling. We find that the truncation of the mGluR1a C-terminal tail prevents mGluR1a phosphorylation and that GRK2 does not contribute to the phosphorylation of an mGluR1 splice variant (mGluR1b). However, mGluR1a-866Delta- and mGluR1b-stimulated inositol phosphate formation is attenuated following GRK2 expression. The expression of the GRK2 C-terminal domain to block membrane translocation of endogenous GRK2 increases mGluR1a-866Delta- and mGluR1b-stimulated inositol phosphate formation, presumably by blocking membrane translocation of GRK2. In contrast, expression of the kinase-deficient GRK2-K220R mutant inhibits inositol phosphate formation by these unphosphorylated receptors. Expression of the GRK2 N-terminal domain (residues 45-185) also attenuates both constitutive and agonist-stimulated mGluR1a, mGluR1a-866Delta, and mGluR1b signaling, and the GRK2 N terminus co-precipitates with mGluR1a. Taken together, our observations indicate that attenuation of mGluR1 signaling by GRK2 is phosphorylation-independent and that the interaction of the N-terminal domain of GRK2 with mGluR1 contributes to the regulation of mGluR1 G protein coupling.


Subject(s)
GTP-Binding Proteins/metabolism , Receptors, Cell Surface/physiology , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction/physiology , Amino Acid Sequence , Cell Line , Humans , Molecular Sequence Data , Mutagenesis , Phosphorylation , Receptors, Metabotropic Glutamate/genetics
17.
Nat Cell Biol ; 4(8): 547-55, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12105416

ABSTRACT

beta-Arrestins are important in chemoattractant receptor-induced granule release, a process that may involve Ral-dependent regulation of the actin cytoskeleton. We have identified the Ral GDP dissociation stimulator (Ral-GDS) as a beta-arrestin-binding protein by yeast two-hybrid screening and co-immunoprecipitation from human polymorphonuclear neutrophilic leukocytes (PMNs). Under basal conditions, Ral-GDS is localized to the cytosol and remains inactive in a complex formed with beta-arrestins. In response to formyl-Met-Leu-Phe (fMLP) receptor stimulation, beta-arrestin Ral-GDS protein complexes dissociate and Ral-GDS translocates with beta-arrestin from the cytosol to the plasma membrane, resulting in the Ras-independent activation of the Ral effector pathway required for cytoskeletal rearrangement. The subsequent re-association of beta-arrestin Ral-GDS complexes is associated with the inactivation of Ral signalling. Thus, beta-arrestins regulate multiple steps in the Ral-dependent processes that result in chemoattractant-induced cytoskeletal reorganization.


Subject(s)
Arrestins/metabolism , Cytoskeleton/metabolism , ral GTP-Binding Proteins/metabolism , ral Guanine Nucleotide Exchange Factor/metabolism , Animals , Arrestins/chemistry , Biological Transport, Active , COS Cells , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cytoskeleton/drug effects , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Macromolecular Substances , Models, Biological , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/drug effects , Neutrophils/metabolism , Rats , Receptors, Formyl Peptide , Receptors, Immunologic/metabolism , Receptors, Peptide/metabolism , Signal Transduction , Two-Hybrid System Techniques , beta-Arrestins , ral GTP-Binding Proteins/chemistry , ral Guanine Nucleotide Exchange Factor/chemistry
18.
J Biol Chem ; 277(1): 679-85, 2002 Jan 04.
Article in English | MEDLINE | ID: mdl-11682489

ABSTRACT

Previous studies have demonstrated that the internalization of the angiotensin II type 1A receptor (AT(1A)R) may be mediated by both beta-arrestin-sensitive and -insensitive mechanisms. Therefore, we have used the AT(1A)R carboxyl-terminal tail to screen a rat brain yeast two-hybrid expression library for novel AT(1A)R-interacting proteins that might contribute to the regulation of AT(1A)R internalization. We have identified Rab5a as an AT(1A)R-binding protein that selectively associates with the AT(1A)R and not with the beta2-adrenergic receptor. A Rab5a-S34N mutant defective in GTP binding does not prevent the internalization of the AT(1A)R but does prevent the trafficking of the AT(1A)R into larger hollow cored vesicular structures. Agonist activation of the AT(1A)R promotes both the formation of Rab5a.AT(1A)R protein complexes and Rab5a GTP binding. Rab5a interactions with the AT(1A)R are mediated in part by the last 10 amino acid residues of the AT(1A)R carboxyl-terminal tail, and although a mutant receptor lacking these residues internalizes normally, it does not redistribute into larger hollow vesicles. Our data suggest that AT(1A)R activation modulates Rab5a activity leading to the homotypic fusion of endocytic vesicles. These observations suggest that vesicular cargo proteins, such as the AT(1A)R, may control their targeting between intracellular compartments by directly regulating the activity of components of the intracellular trafficking machinery such as Rab5a.


Subject(s)
Guanosine Triphosphate/metabolism , Membrane Fusion , Receptors, Angiotensin/metabolism , Transport Vesicles/metabolism , rab5 GTP-Binding Proteins/metabolism , Amino Acid Sequence , Animals , COS Cells , ErbB Receptors/physiology , Molecular Sequence Data , Receptor, Angiotensin, Type 1 , Receptors, Angiotensin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...