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










Database
Language
Publication year range
1.
J Cell Physiol ; 231(12): 2768-78, 2016 12.
Article in English | MEDLINE | ID: mdl-27017909

ABSTRACT

Proper regulation of cytosolic Ca(2+) is critical for pancreatic acinar cell function. Disruptions in normal Ca(2+) concentrations affect numerous cellular functions and are associated with pancreatitis. Membrane pumps and channels regulate cytosolic Ca(2+) homeostasis by promoting rapid Ca(2+) movement. Determining how expression of Ca(2+) modulators is regulated and the cellular alterations that occur upon changes in expression can provide insight into initiating events of pancreatitis. The goal of this study was to delineate the gene structure and regulation of a novel pancreas-specific isoform for Secretory Pathway Ca(2+) ATPase 2 (termed SPCA2C), which is encoded from the Atp2c2 gene. Using Next Generation Sequencing of RNA (RNA-seq), chromatin immunoprecipitation for epigenetic modifications and promoter-reporter assays, a novel transcriptional start site was identified that promotes expression of a transcript containing the last four exons of the Atp2c2 gene (Atp2c2c). This region was enriched for epigenetic marks and pancreatic transcription factors that promote gene activation. Promoter activity for regions upstream of the ATG codon in Atp2c2's 24th exon was observed in vitro but not in in vivo. Translation from this ATG encodes a protein aligned with the carboxy terminal of SPCA2. Functional analysis in HEK 293A cells indicates a unique role for SPCA2C in increasing cytosolic Ca(2+) . RNA analysis indicates that the decreased Atp2c2c expression observed early in experimental pancreatitis reflects a global molecular response of acinar cells to reduce cytosolic Ca(2+) levels. Combined, these results suggest SPCA2C affects Ca(2+) homeostasis in pancreatic acinar cells in a unique fashion relative to other Ca(2+) ATPases. J. Cell. Physiol. 231: 2768-2778, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Acinar Cells/metabolism , Calcium-Transporting ATPases/genetics , Pancreas/pathology , Transcription Initiation Site , Transcription, Genetic , Acinar Cells/pathology , Amino Acid Sequence , Animals , Base Sequence , Calcium/metabolism , Calcium-Transporting ATPases/chemistry , Calcium-Transporting ATPases/metabolism , Ceruletide , Epigenesis, Genetic , Exons/genetics , Female , Genome , HEK293 Cells , Histones/metabolism , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Pancreatitis/genetics , Pancreatitis/pathology , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism
2.
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
3.
J Neurosci ; 30(1): 316-24, 2010 Jan 06.
Article in English | MEDLINE | ID: mdl-20053912

ABSTRACT

Huntington's disease (HD) is an autosomal-dominant neurodegenerative disorder caused by a polyglutamine expansion in the huntingtin protein (Htt). Group I metabotropic glutamate receptors (mGluRs) are coupled to G(alphaq) and play an important role in neuronal survival. We have previously demonstrated that mGluRs interact with Htt. Here we used striatal neuronal primary cultures and acute striatal slices to demonstrate that mGluR-mediated signaling pathways are altered in a presymptomatic mouse model of HD (Hdh(Q111/Q111)), as compared to those of control mice (Hdh(Q20/Q20)). mGluR1/5-mediated inositol phosphate (InsP) formation is desensitized in striatal slices from Hdh(Q111/Q111) mice and this desensitization is PKC-mediated. Despite of decreased InsP formation, (S)-3,5-dihydroxylphenylglycine (DHPG)-mediated Ca(2+) release is higher in Hdh(Q111/Q111) than in Hdh(Q20/Q20) neurons. Furthermore, mGluR1/5-stimulated AKT and extracellular signal-regulated kinase (ERK) activation is altered in Hdh(Q111/Q111) mice. Basal AKT activation is higher in Hdh(Q111/Q111) neurons and this increase is mGluR5 dependent. Moreover, mGluR5 activation leads to higher levels of ERK activation in Hdh(Q111/Q111) than in Hdh(Q20/Q20) striatum. PKC inhibition not only brings Hdh(Q111/Q111) DHPG-stimulated InsP formation to Hdh(Q20/Q20) levels, but also causes an increase in neuronal cell death in Hdh(Q111/Q111) neurons. However, PKC inhibition does not modify neuronal cell death in Hdh(Q20/Q20) neurons, suggesting that PKC-mediated desensitization of mGluR1/5 in Hdh(Q111/Q111) mice might be protective in HD. Together, these data indicate that group I mGluR-mediated signaling pathways are altered in HD and that these cell signaling adaptations could be important for striatal neurons survival.


Subject(s)
Disease Models, Animal , Huntington Disease/physiopathology , Receptors, Metabotropic Glutamate/physiology , Signal Transduction/physiology , Animals , Cells, Cultured , Gene Knock-In Techniques , Mice , Mice, Transgenic
4.
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
5.
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
6.
J Biol Chem ; 284(35): 23444-53, 2009 Aug 28.
Article in English | MEDLINE | ID: mdl-19564331

ABSTRACT

The uncoupling of metabotropic glutamate receptors (mGluRs) from heterotrimeric G proteins represents an essential feedback mechanism that protects neurons against receptor overstimulation that may ultimately result in damage. The desensitization of mGluR signaling is mediated by both second messenger-dependent protein kinases and G protein-coupled receptor kinases (GRKs). Unlike mGluR1, the attenuation of mGluR5 signaling in HEK 293 cells is reported to be mediated by a phosphorylation-dependent mechanism. However, the mechanisms regulating mGluR5 signaling and endocytosis in neurons have not been investigated. Here we show that a 2-fold overexpression of GRK2 leads to the attenuation of endogenous mGluR5-mediated inositol phosphate (InsP) formation in striatal neurons and siRNA knockdown of GRK2 expression leads to enhanced mGluR5-mediated InsP formation. Expression of a catalytically inactive GRK2-K220R mutant also effectively attenuates mGluR5 signaling, but the expression of a GRK2-D110A mutant devoid in Galpha(q/11) binding increases mGluR5 signaling in response to agonist stimulation. Taken together, these results indicate that the attenuation of mGluR5 responses in striatal neurons is phosphorylation-independent. In addition, we find that mGluR5 does not internalize in response to agonist treatment in striatal neuron, but is efficiently internalized in cortical neurons that have higher levels of endogenous GRK2 protein expression. When overexpressed in striatal neurons, GRK2 promotes agonist-stimulated mGluR5 internalization. Moreover, GRK2-mediated promotion of mGluR5 endocytosis does not require GRK2 catalytic activity. Thus, we provide evidence that GRK2 mediates phosphorylation-independent mGluR5 desensitization and internalization in neurons.


Subject(s)
Endocytosis , G-Protein-Coupled Receptor Kinase 2/metabolism , Neurons/physiology , Receptors, Metabotropic Glutamate/metabolism , Animals , Cells, Cultured , G-Protein-Coupled Receptor Kinase 2/genetics , Inositol Phosphates/metabolism , Mice , Mice, Knockout , Neurons/enzymology , Phosphorylation , Protein Binding , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/genetics
7.
Neurochem Int ; 50(2): 356-64, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17092608

ABSTRACT

Trafficking of the vesicular acetylcholine transporter (VAChT) to synaptic vesicles has the potential to regulate storage and release of acetylcholine. We used the C-terminal tail of the vesicular acetylcholine transporter as bait for the screening of a brain cDNA library by yeast-two hybrids. Here we report an interaction uncovered in this screening with SEC14L1, a mammalian SEC14-like protein that may function as a phospholipid transfer protein. The interaction of VAChT and SEC14L1 occurred through the GOLD domain found in the latter and was confirmed in mammalian cells. In addition, we also found that SEC14L1 co-immunoprecipitates with the high affinity choline transporter (CHT1), but not with synaptophysin or synaptotagmin. In cultured cells SEC14L1 was predominantly found in the cytosol with little or no localization in defined organelles. In contrast, overexpression of VAChT or CHT1 with SEC14L1 recruited the latter to large intracellular organelles similar to vesicles or vesicle aggregates. Finally, we find that overexpression of SEC14L1 modestly decreases high affinity choline transport activity. We suggest that interaction of cholinergic transporters with proteins containing the GOLD domain may be relevant for transporter function.


Subject(s)
Carrier Proteins/metabolism , Lipoproteins/metabolism , Trans-Activators/metabolism , Vesicular Acetylcholine Transport Proteins/metabolism , Amino Acid Sequence , Animals , Brain Chemistry/genetics , Cation Transport Proteins/metabolism , Cell Line , Cells, Cultured , Choline/metabolism , Cloning, Molecular , Cytosol/metabolism , DNA, Complementary/genetics , Fluorescent Antibody Technique , Gene Library , Humans , Immunoprecipitation , Microscopy, Confocal , Molecular Sequence Data , PC12 Cells , Phospholipid Transfer Proteins/metabolism , Plasmids/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Synaptic Transmission/physiology , Synaptophysin/metabolism , Synaptotagmins/metabolism , Transfection
8.
J Neurochem ; 94(4): 957-69, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16092939

ABSTRACT

The vesicular acetylcholine transporter (VAChT) regulates the amount of acetylcholine stored in synaptic vesicles. However, the mechanisms that control the targeting of VAChT and other synaptic vesicle proteins are still poorly comprehended. These processes are likely to depend, at least partially, on structural determinants present in the primary sequence of the protein. Here, we use site-directed mutagenesis to evaluate the contribution of the C-terminal tail of VAChT to the targeting of this transporter to synaptic-like microvesicles in cholinergic SN56 cells. We found that residues 481-490 contain the trafficking information necessary for VAChT localization and that within this region L485 and L486 are strictly necessary. Deletion and alanine-scanning mutants lacking most of the carboxyl tail of VAChT, but containing residues 481-490, were still targeted to microvesicles. Moreover, we found that clathrin-mediated endocytosis of VAChT is required for targeting to microvesicles in SN56 and PC12 cells. The data provide novel information on the mechanisms and structural determinants necessary for VAChT localization to synaptic vesicles.


Subject(s)
Homeostasis , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Synaptic Vesicles/metabolism , Amino Acid Sequence , Animals , Cell Line , Clathrin/physiology , Endocytosis/physiology , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , PC12 Cells , Protein Conformation , Rats , Tissue Distribution , Vesicular Acetylcholine Transport Proteins
9.
J Neurochem ; 82(5): 1221-8, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12358769

ABSTRACT

The pathways by which synaptic vesicle proteins reach their destination are not completely defined. Here we investigated the traffic of a green fluorescent protein (GFP)-tagged version of the vesicular acetylcholine transporter (VAChT) in cholinergic SN56 cells, a model system for neuronal processing of this cargo. GFP-VAChT accumulates in small vesicular compartments in varicosities, but perturbation of endocytosis with a dominant negative mutant of dynamin I-K44A impaired GFP-VAChT trafficking to these processes. The protein in this condition accumulated in the cell body plasma membrane and in large vesicular patches therein. A VAChT endocytic mutant (L485A/L486A) was also located at the plasma membrane, however, the protein was not sorted to dynamin I-K44A generated vesicles. A fusion protein containing the VAChT C-terminal tail precipitated the AP-2 adaptor protein complex from rat brain, suggesting that VAChT directly interacts with the endocytic complex. In addition, yeast two hybrid experiments indicated that the C-terminal tail of VAChT interacts with the micro subunit of AP-2 in a di-leucine (L485A/L486A) dependent fashion. These observations suggest that the di-leucine motif regulates sorting of VAChT from the soma plasma membrane through a clathrin dependent mechanism prior to the targeting of the transporter to varicosities.


Subject(s)
Carrier Proteins/metabolism , GTP Phosphohydrolases/metabolism , Membrane Proteins/metabolism , Membrane Transport Proteins , Neurons/metabolism , Vesicular Transport Proteins , Adaptor Proteins, Vesicular Transport , Amino Acid Motifs/physiology , Amino Acid Substitution , Animals , Carrier Proteins/genetics , Cell Line , Dynamin I , Dynamins , Endocytosis/drug effects , Endocytosis/physiology , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/pharmacology , Genes, Dominant , Green Fluorescent Proteins , Luminescent Proteins/genetics , Macromolecular Substances , Mice , Neurons/cytology , Protein Binding/physiology , Protein Transport/physiology , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection , Two-Hybrid System Techniques , Vesicular Acetylcholine Transport Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...