Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Mol Cell Res ; 1866(8): 1249-1259, 2019 08.
Article in English | MEDLINE | ID: mdl-30951783

ABSTRACT

Intact store-operated calcium entry (SOCE) mechanisms ensure the maintenance of Ca2+ homeostasis in cardiomyocytes while their dysregulation promotes the development of cardiomyopathies. To better understand this calcium handling process in cardiomyocytes, we sought to identify unknown protein partners of stromal interaction molecule 1 (STIM1), a main regulatory protein of SOCE. We identified the muscle-related coiled-coil protein (MURC), also known as Cavin-4, as a candidate and showed that MURC interacts with STIM1 in cardiomyocytes. This interaction occurs via the HR1 and ERM domains of MURC and STIM1, respectively. Our results also demonstrated that the overexpression of MURC in neonatal rat ventricular myocytes (NRVM) is sufficient to potentiate SOCE and that its HR1 domain is required to mediate this effect. Interestingly, the R140W-MURC mutant, a missense variant of the HR1 domain associated with human dilated cardiomyopathy, exacerbates the SOCE increase in NRVM. Although the endogenous expression of STIM1 and Ca2+ channel Orai1 is not modulated under these conditions, we showed that MURC increases the interaction between these proteins under resting conditions. Our study provides novel evidence that MURC regulates SOCE by interacting with STIM1 in cardiomyocytes. In addition, we identified a first potential mechanism by which the R140W mutation of MURC may contribute to calcium mishandling and the development of cardiomyopathies.


Subject(s)
Calcium Signaling , Calcium/metabolism , Cardiomyopathy, Dilated/metabolism , Muscle Proteins/metabolism , Myocytes, Cardiac/metabolism , Vesicular Transport Proteins/metabolism , Amino Acid Substitution , Animals , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Cells, Cultured , Humans , Muscle Proteins/genetics , Mutation, Missense , Myocytes, Cardiac/pathology , ORAI1 Protein/genetics , ORAI1 Protein/metabolism , Protein Domains , Rats , Rats, Sprague-Dawley , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , Vesicular Transport Proteins/genetics
2.
Front Immunol ; 8: 94, 2017.
Article in English | MEDLINE | ID: mdl-28223986

ABSTRACT

T lymphocytes from Gimap5lyp/lyp rats carrying a recessive mutation in the GTPase of immune-associated protein 5 (Gimap5) gene undergo spontaneous apoptosis. Molecular mechanisms underlying this survival defect are not yet clear. We have shown that Gimap5lyp/lyp T lymphocytes display reduced calcium influx following T cell antigen receptor (TCR) stimulation that was associated with impaired buffering of calcium by mitochondria. Here, we investigated the subcellular localization of GIMAP5 and its influence on Ca2+ response in HEK293T cells and T lymphocytes. The more abundantly expressed GIMAP5v2 localizes to the lysosome and certain endosomal vesicles. Gimap5lyp/lyp T lymphocytes showed increased accumulation of calcium in the lysosomes as evidenced by Gly-Phe ß-naphthylamide (GPN) triggered Ca2+ release. As a corollary, GPN-induced Ca2+ flux was decreased in HEK293T cells expressing GIMAP5v2. Strikingly, TCR stimulation of rat, mouse, and human T lymphocytes increased lysosomal calcium content. Overall, our findings show that lysosomes modulate cellular Ca2+ response during T cell activation and that GIMAP5 regulates the lysosomal Ca2+ compartment in T lymphocytes.

3.
Metallomics ; 6(7): 1269-76, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24733507

ABSTRACT

The canonical transient receptor potential 6 (TRPC6) protein is a non-selective cation channel able to transport essential trace elements like iron (Fe) and zinc (Zn) through the plasma membrane. Its over-expression in HEK-293 cells causes an intracellular accumulation of Zn, indicating that it could be involved in Zn transport. This finding prompted us to better understand the role played by TRPC6 in Zn homeostasis. Experiments done using the fluorescent probe FluoZin-3 showed that HEK cells possess an intracellular pool of mobilisable Zn present in compartments sensitive to the vesicular proton pump inhibitor Baf-A, which affects endo/lysosomes. TRPC6 over-expression facilitates the basal uptake of Zn and enhances the size of the pool of Zn sensitive to Baf-A. Quantitative RT-PCR experiments showed that TRPC6 over-expression does not affect the mRNA expression of Zn transporters (ZnT-1, ZnT-5, ZnT-6, ZnT-7, ZnT-9, Zip1, Zip6, Zip7, and Zip14); however it up-regulates the mRNA expression of metallothionein-I and -II. This alters the Zn buffering capacities of the cells as illustrated by the experiments done using the Zn ionophore Na pyrithione. In addition, HEK cells over-expressing TRPC6 grow slower than their parental HEK cells. This feature can be mimicked by growing HEK cells in a culture medium supplemented with 5 µM of Zn acetate. Finally, a proteomic analysis revealed that TRPC6 up-regulates the expression of the actin-associated proteins ezrin and cofilin-1, and changes the organisation of the actin cytoskeleton without changing the cellular actin content. Altogether, these data indicate that TRPC6 is participating in the transport of Zn and influences the Zn storage and buffering capacities of the cells.


Subject(s)
TRPC Cation Channels/biosynthesis , Zinc/metabolism , Actin Depolymerizing Factors/biosynthesis , Cation Transport Proteins/metabolism , Cytoskeletal Proteins/biosynthesis , HEK293 Cells , Homeostasis/drug effects , Humans , Polycyclic Compounds/pharmacology , Proton Pump Inhibitors/pharmacology , TRPC6 Cation Channel
4.
Biochim Biophys Acta ; 1833(6): 1294-303, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23458835

ABSTRACT

STIM1 plays a crucial role in Ca(2+) homeostasis, particularly in replenishing the intracellular Ca(2+) store following its depletion. In cardiomyocytes, the Ca(2+) content of the sarcoplasmic reticulum must be tightly controlled to sustain contractile activity. The presence of STIM1 in cardiomyocytes suggests that it may play a role in regulating the contraction of cardiomyocytes. The aim of the present study was to determine how STIM1 participates in the regulation of cardiac contractility. Atomic force microscopy revealed that knocking down STIM1 disrupts the contractility of cardiomyocyte-derived HL-1 cells. Ca(2+) imaging also revealed that knocking down STIM1 causes irregular spontaneous Ca(2+) oscillations in HL-1 cells. Action potential recordings further showed that knocking down STIM1 induces early and delayed afterdepolarizations. Knocking down STIM1 increased the peak amplitude and current density of T-type voltage-dependent Ca(2+) channels (T-VDCC) and shifted the activation curve toward more negative membrane potentials in HL-1 cells. Biotinylation assays revealed that knocking down STIM1 increased T-VDCC surface expression and co-immunoprecipitation assays suggested that STIM1 directly regulates T-VDCC activity. Thus, STIM1 is a negative regulator of T-VDCC activity and maintains a constant cardiac rhythm by preventing a Ca(2+) overload that elicits arrhythmogenic events.


Subject(s)
Calcium Channels, T-Type/metabolism , Calcium Signaling/physiology , Calcium/metabolism , Membrane Glycoproteins/metabolism , Muscle Contraction , Myocytes, Cardiac/metabolism , Tachycardia , Animals , Blotting, Western , Calcium Channels , Cells, Cultured , Electrophysiology , Immunoprecipitation , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Mice , Myocytes, Cardiac/cytology , Stromal Interaction Molecule 1
5.
Biochem J ; 449(2): 353-64, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23098229

ABSTRACT

Mature T-lymphocytes undergo spontaneous apoptosis in the biobreeding diabetes-prone strain of rats due to the loss of the functional GIMAP5 (GTPase of the immune-associated nucleotide-binding protein 5) protein. The mechanisms underlying the pro-survival function of GIMAP5 in T-cells have not yet been elucidated. We have previously shown that GIMAP5 deficiency in T-cells impairs Ca2+ entry via plasma membrane channels following exposure to thapsigargin or stimulation of the T-cell antigen receptor. In the present study we report that this reduced Ca2+ influx in GIMAP5-deficient T-cells is associated with the inability of their mitochondria to sequester Ca2+ following capacitative entry, which is required for sustained Ca2+ influx via the plasma membrane channels. Consistent with a role for GIMAP5 in regulating mitochondrial Ca2+, overexpression of GIMAP5 in HEK (human embryonic kidney)-293 cells resulted in increased Ca2+ accumulation within the mitochondria. Disruption of microtubules, but not the actin cytoskeleton, abrogated mitochondrial Ca2+ sequestration in primary rat T-cells, whereas both microtubules and actin cytoskeleton were needed for the GIMAP5-mediated increase in mitochondrial Ca2+ in HEK-293 cells. Moreover, GIMAP5 showed partial colocalization with tubulin in HEK-293 cells. On the basis of these findings, we propose that the pro-survival function of GIMAP5 in T-lymphocytes may be linked to its requirement to facilitate microtubule-dependent mitochondrial buffering of Ca2+ following capacitative entry.


Subject(s)
Calcium/metabolism , GTP-Binding Proteins/metabolism , Mitochondria/metabolism , T-Lymphocytes/metabolism , Animals , Blotting, Western , Cell Membrane/metabolism , Cells, Cultured , Cytoskeleton/metabolism , GTP-Binding Proteins/genetics , HEK293 Cells , Humans , Ion Transport , Microscopy, Confocal , Mutation , Rats , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/cytology , Tubulin/metabolism
6.
J Biol Chem ; 287(21): 17672-17681, 2012 May 18.
Article in English | MEDLINE | ID: mdl-22493444

ABSTRACT

TRPC6 is a cation channel in the plasma membrane that plays a role in Ca(2+) entry after the stimulation of a G(q)-protein-coupled or tyrosine-kinase receptor. TRPC6 translocates to the plasma membrane upon stimulation and remains there as long as the stimulus is present. However, the mechanism that regulates the trafficking and activation of TRPC6 are unclear. In this study we showed phosphoinositide 3-kinase and its antagonistic phosphatase, PTEN, are involved in the activation of TRPC6. The inhibition of PI3K by PIK-93, LY294002, or wortmannin decreased carbachol-induced translocation of TRPC6 to the plasma membrane and carbachol-induced net Ca(2+) entry into T6.11 cells. Conversely, a reduction of PTEN expression did not affect carbachol-induced externalization of TRPC6 but increased Ca(2+) entry through TRPC6 in T6.11 cells. We also showed that the PI3K/PTEN pathway regulates vasopressin-induced translocation of TRPC6 to the plasma membrane and vasopressin-induced Ca(2+) entry into A7r5 cells, which endogenously express TRPC6. In summary, we provided evidence that the PI3K/PTEN pathway plays an important role in the translocation of TRPC6 to the plasma membrane and may thus have a significant impact on Ca(2+) signaling in cells that endogenously express TRPC6.


Subject(s)
Calcium Signaling/physiology , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , TRPC Cation Channels/metabolism , Androstadienes/pharmacology , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Carbachol/pharmacology , Cardiotonic Agents/pharmacology , Cell Membrane/genetics , Cell Membrane/metabolism , Chromones/pharmacology , HEK293 Cells , Humans , Mice , Morpholines/pharmacology , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/genetics , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Transport/drug effects , Protein Transport/physiology , TRPC Cation Channels/genetics , TRPC6 Cation Channel , Wortmannin
7.
Biochim Biophys Acta ; 1808(12): 2807-18, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21864503

ABSTRACT

TRPC6 are plasma membrane cation channels. By means of live-cell imaging and spectroscopic methods, we found that HEK cells expressing TRPC6 channels (HEK-TRPC6) are enriched in zinc and sulphur and have a reduced copper content when compared to HEK cells and HEK cells expressing TRPC3 channels (HEK-TRPC3). Hence, HEK-TRPC6 cells have larger pools of mobilizable Zn2+ and are more sensitive to an oxidative stress. Synchrotron X-ray fluorescence experiments showed a higher zinc content in the nuclear region indicating that the intracellular distribution of this metal was influenced by the over-expression of TRPC6 channels. Their properties were investigated with the diacylglycerol analogue SAG and the plant extract hyperforin. Electrophysiological recordings and imaging experiments with the fluorescent Zn2+ probe FluoZin-3 demonstrated that TRPC6 channels form Zn2+-conducting channels. In cortical neurons, hyperforin-sensitive channels co-exist with voltage-gated channels, AMPA and NMDA receptors, which are known to transport Zn2+. The ability of these channels to regulate the size of the mobilizable pools of Zn2+ was compared. The data collected indicate that the entry of Zn2+ through TRPC6 channels can up-regulate the size of the DTDP-sensitive pool of Zn2+. By showing that TRPC6 channels constitute a Zn2+ entry pathway, our study suggests that they could play a role in zinc homeostasis.


Subject(s)
TRPC Cation Channels/metabolism , Zinc/metabolism , Cell Line , Colorimetry , Fluorescent Dyes/metabolism , Homeostasis , Humans , TRPC6 Cation Channel
8.
PLoS One ; 6(3): e18121, 2011 Mar 23.
Article in English | MEDLINE | ID: mdl-21448286

ABSTRACT

TRPC are nonselective cation channels involved in calcium entry. Their regulation by phosphorylation has been shown to modulate their routing and activity. TRPC6 activity increases following phosphorylation by Fyn, and is inhibited by protein kinase G and protein kinase C. A previous study by our group showed that TRPC6 is phosphorylated under unstimulated conditions in a human embryonic kidney cells line (HEK293). To investigate the mechanism responsible for this phosphorylation, we used a MS/MS approach combined with metabolic labeling and showed that the serine at position 814 is phosphorylated in unstimulated cells. The mutation of Ser(814) into Ala decreased basal phosphorylation but did not modify TRPC6 activity. Even though Ser(814) is within a consensus site for casein kinase II (CK2), we showed that CK2 is not involved in the phosphorylation of TRPC6 and does not modify its activity. In summary, we identified a new basal phosphorylation site (Ser(814)) on TRPC6 and showed that CK2 is not responsible for the phosphorylation of this site.


Subject(s)
Phosphoserine/metabolism , TRPC Cation Channels/metabolism , Amino Acid Sequence , Amino Acid Substitution/genetics , Animals , Casein Kinase II/antagonists & inhibitors , HEK293 Cells , Humans , Mass Spectrometry , Mice , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutation/genetics , Phosphorylation/drug effects , Phosphoserine/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Transport/drug effects , Staining and Labeling , TRPC6 Cation Channel
9.
Proc Natl Acad Sci U S A ; 108(4): 1693-8, 2011 Jan 25.
Article in English | MEDLINE | ID: mdl-21220323

ABSTRACT

Insulin secretion by pancreatic ß cells is a complex and highly regulated process. Disruption of this process can lead to diabetes mellitus. One of the various pathways involved in the regulation of insulin secretion is the activation of heterotrimeric G proteins. Bordetella pertussis toxin (PTX) promotes insulin secretion, suggesting the involvement of one or more of three G(i) and/or two G(o) proteins as suppressors of insulin secretion from ß cells. However, neither the mechanism of this inhibitory modulation of insulin secretion nor the identity of the G(i/o) proteins involved has been elucidated. Here we show that one of the two splice variants of G(o), G(o2), is a key player in the control of glucose-induced insulin secretion by ß cells. Mice lacking G(o2)α, but not those lacking α subunits of either G(o1) or any G(i) proteins, handle glucose loads more efficiently than wild-type (WT) mice, and do so by increased glucose-induced insulin secretion. We thus provide unique genetic evidence that the G(o2) protein is a transducer in an inhibitory pathway that prevents damaging oversecretion of insulin.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Glucose/pharmacology , Insulin/metabolism , Islets of Langerhans/drug effects , Animals , Blotting, Western , Brain/metabolism , Female , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Gene Expression , Glucose Tolerance Test , Immunohistochemistry , In Vitro Techniques , Insulin/genetics , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Isoforms/genetics , Protein Isoforms/metabolism , Reverse Transcriptase Polymerase Chain Reaction
10.
J Biol Chem ; 285(52): 40534-43, 2010 Dec 24.
Article in English | MEDLINE | ID: mdl-20961851

ABSTRACT

TRPC6 is a cation channel in the plasma membrane that plays a role in Ca(2+) entry following the stimulation of a G(q)-protein coupled or tyrosine kinase receptor. A dysregulation of TRPC6 activity causes abnormal proliferation of smooth muscle cells and glomerulosclerosis. In the present study, we investigated the regulation of TRPC6 activity by protein kinase C (PKC). We showed that inhibiting PKC with GF1 or activating it with phorbol 12-myristate 13-acetate potentiated and inhibited agonist-induced Ca(2+) entry, respectively, into cells expressing TRPC6. Similar results were obtained when TRPC6 was directly activated with 1-oleyl-2-acetyl-sn-glycerol. Activation of the cells with carbachol increased the phosphorylation of TRPC6, an effect that was prevented by the inhibition of PKC. The target residue of PKC was identified by an alanine screen of all canonical PKC sites on TRPC6. Unexpectedly, all the mutants, including TRPC6(S768A) (a residue previously proposed to be a target for PKC), displayed PKC-dependent inhibition of channel activity. Phosphorylation prediction software suggested that Ser(448), in a non-canonical PKC consensus sequence, was a potential target for PKCδ. Ba(2+) and Ca(2+) entry experiments revealed that GF1 did not potentiate TRPC6(S448A) activity. Moreover, activation of PKC did not enhance the phosphorylation state of TRPC6(S448A). Using A7r5 vascular smooth muscle cells, which endogenously express TRPC6, we observed that a novel PKC isoform is involved in the inhibition of the vasopressin-induced Ca(2+) entry. Furthermore, knocking down PKCδ in A7r5 cells potentiated vasopressin-induced Ca(2+) entry. In summary, we provide evidence that PKCδ exerts a negative feedback effect on TRPC6 through the phosphorylation of Ser(448).


Subject(s)
Myocytes, Smooth Muscle/metabolism , Protein Kinase C-delta/metabolism , TRPC Cation Channels/metabolism , Amino Acid Substitution , Carbachol/pharmacology , Carcinogens/pharmacology , Cell Proliferation/drug effects , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/metabolism , HEK293 Cells , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Miotics/pharmacology , Mutation, Missense , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase C-delta/genetics , Serine/genetics , Serine/metabolism , TRPC Cation Channels/genetics , TRPC6 Cation Channel , Tetradecanoylphorbol Acetate/pharmacology , Vasoconstrictor Agents/pharmacology , Vasopressins/pharmacology
11.
Diabetes ; 59(10): 2522-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20622165

ABSTRACT

OBJECTIVE: Pertussis toxin uncoupling-based studies have shown that Gαi and Gαo can inhibit insulin secretion in pancreatic ß-cells. Yet it is unclear whether Gαi and Gαo operate through identical mechanisms and how these G-protein-mediated signals inhibit insulin secretion in vivo. Our objective is to examine whether/how Gαo regulates islet development and insulin secretion in ß-cells. RESEARCH DESIGN AND METHODS: Immunoassays were used to analyze the Gαo expression in mouse pancreatic cells. Gαo was specifically inactivated in pancreatic progenitor cells by pancreatic cell-specific gene deletion. Hormone expression and insulin secretion in response to different stimuli were assayed in vivo and in vitro. Electron microscope and total internal reflection fluorescence-based assays were used to evaluate how Gαo regulates insulin vesicle docking and secretion in response to glucose stimulation. RESULTS: Islet cells differentiate properly in Gαo(-/-) mutant mice. Gαo inactivation significantly enhances insulin secretion both in vivo and in isolation. Gαo nullizygous ß-cells contain an increased number of insulin granules docked on the cell plasma membrane, although the total number of vesicles per ß-cell remains unchanged. CONCLUSIONS: Gαo is not required for endocrine islet cell differentiation, but it regulates the number of insulin vesicles docked on the ß-cell membrane.


Subject(s)
GTP-Binding Protein alpha Subunits/antagonists & inhibitors , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Animals , Cell Differentiation , GTP-Binding Protein alpha Subunits/genetics , Gene Expression Regulation , Homeodomain Proteins/genetics , Homeostasis , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/physiology , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Trans-Activators/genetics
12.
J Cell Physiol ; 225(2): 444-53, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20458742

ABSTRACT

Although most signaling responses initiated by tumor necrosis factor-alpha (TNF-alpha) occur in a Ca(2+)-independent fashion, TNF-alpha receptor signaling augments Ca(2+) entry induced by Galpha(q/11) G-protein coupled receptors (GPCRs) in endothelial cells and increases trans-endothelial permeability. The signaling events involved in GPCR-induced Ca(2+) influx have been characterized and involve store-operated Ca(2+) entry facilitated by the Ca(2+) permeable ion channel, transient receptor potential canonical 4 (TRPC4). Little is known about the mechanisms by which TNF-alpha receptor signaling augments GPCR-induced Ca(2+) entry. TNF-alpha Receptor Ubiquitous Signaling and Scaffolding protein (TRUSS) is a tumor necrosis factor receptor-1 (TNF-R1)-associated protein whose gene name is TRPC4-associated protein (TRPC4AP). The goal of our study was to test the hypothesis that TRUSS serves to link TNF-R1 and GPCR-signaling pathways at the level of TRPC4 by: (i) determining if TRUSS and TNF-R1 interact with TRPC4, and (ii) investigating the role of TRUSS, TNF-R1, and TRPC4 in GPCR-induced Ca(2+) signaling. Here, we show that TRUSS and TNF-R1 interact with a sub-family of TRPC channels (TRPC1, 4, and 5). In addition, we show that TRUSS and TNF-R1 function together with TRPC4 to elevate endoplasmic reticulum Ca(2+) filling in the context of reduced endoplasmic reticulum Ca(2+) storage initiated by G-protein coupled m1 muscarinic acetylcholine receptor (m1AchR) signaling. Together, these findings suggest that TNF-R1, TRUSS, and TRPC4 augment Ca(2+) loading of endoplasmic reticulum Ca(2+) stores in the context of m1AchR stimulation and provide new insights into the mechanisms that connect TNF-R1 to GPCR-induced Ca(2+) signaling.


Subject(s)
Calcium/metabolism , Receptor, Muscarinic M1/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction/physiology , TRPC Cation Channels/metabolism , Cell Line , Cell Membrane/physiology , Endoplasmic Reticulum/metabolism , Gene Expression Regulation/physiology , Humans , Permeability , Receptor, Muscarinic M1/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , TRPC Cation Channels/genetics
13.
Biochim Biophys Acta ; 1803(7): 805-12, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20346379

ABSTRACT

TRPC proteins become involved in Ca2+ entry following the activation of Gq-protein coupled receptors. TRPC6 is inserted into the plasma membrane upon stimulation and remains in the plasma membrane as long as the stimulus is present. However, the mechanism that regulates the trafficking of TRPC6 is unclear. In the present study, we highlighted the involvement of two Rab GTPases in the trafficking of TRPC6. Rab9 co-localized in vesicular structures with TRPC6 in HeLa cells and co-immunoprecipitated with TRPC6. When co-expressed with TRPC6, Rab9(S21N), a dominant negative mutant, caused an increase in the level of TRPC6 at the plasma membrane and in TRPC6-mediated Ca2+ entry upon activation by a muscarinic receptor agonist. Similarly, the expression of Rab11 also caused an increase in TRPC6 expression at the cell surface and an increase in TRPC6-mediated Ca2+ entry. The co-expression of TRPC6 with the dominant negative mutant Rab11(S25N) abolished CCh-induced TRPC6 activation and reduced the level of TRPC6 at the plasma membrane. This study demonstrates that the trans-Golgi network and recycling endosomes are involved in the intracellular trafficking of TRPC6 by regulating channel density at the cell surface.


Subject(s)
TRPC Cation Channels/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Biological Transport/physiology , Cell Membrane/metabolism , HeLa Cells , Humans , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , TRPC Cation Channels/genetics , TRPC6 Cation Channel , rab GTP-Binding Proteins/genetics
14.
J Immunol ; 183(4): 2747-57, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19620302

ABSTRACT

Certain proteins, including receptors and signaling molecules, are known to be enriched in caveolae and lipid rafts. Caveolin-1, the major structural protein of caveolae, specifically interacts with many signaling molecules and, thus, caveolae and lipid rafts are often seen as preassembled signaling platforms. A potential binding site for caveolin-1 is present in the platelet-activating factor receptor (PAFR) sequence, and many downstream signaling components of PAFR activation preferentially localize in caveolae. The aim of this study was to investigate whether the PAFR was localized in caveolae/lipid raft domains and, if so, what would be the significance of such localization for PAFR signaling. In this study, we demonstrate that PAFR localizes within membrane microdomains, in close proximity to caveolin-1 in living cells, with potential interaction through a caveolin-1-binding sequence in the PAFR C terminus. Caveolin-1, however, is not essential for PAFR localization in lipid rafts. Disruption of caveolae/lipid rafts with methyl-beta-cyclodextrin markedly reduced PAF-triggered inositol phosphate production and cytosolic calcium flux, suggesting that PAFR signaling through the Galphaq protein was critically dependent on integrity of lipid rafts and/or caveolae. Interestingly, whereas in caveolin-1-expressing cells lipid raft disruption markedly decreased PAFR-mediated activation of the ERK/MAPK pathway, in cells lacking caveolae, such as leukocytes, lipid raft disruption had either the same inhibitory effect (Ramos B cells) or no effect (monocytes) on PAFR capacity to signal through the ERK/MAPK pathway. In conclusion, PAFR appears to localize within caveolae or lipid rafts in different cell types, and this location may be important for specific signaling events.


Subject(s)
Calcium Signaling , Caveolae/metabolism , Caveolin 1/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Platelet Activating Factor/physiology , Animals , Binding Sites/immunology , CHO Cells , Calcium Signaling/immunology , Caveolae/enzymology , Caveolae/immunology , Cell Line , Cell Line, Tumor , Cricetinae , Cricetulus , Enzyme Activation/immunology , Humans , Membrane Microdomains/immunology , Membrane Microdomains/metabolism , Peptide Fragments/metabolism , Platelet Membrane Glycoproteins/metabolism , Platelet Membrane Glycoproteins/physiology , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/physiology
15.
Endocrinology ; 150(5): 2190-6, 2009 May.
Article in English | MEDLINE | ID: mdl-19147678

ABSTRACT

The inositol 1,4,5-trisphosphate receptor (IP(3)R) is a Ca(2+) release channel that plays a pivotal role in regulating intracellular Ca(2+) levels in resting cells. Three isoforms of IP(3)Rs have been identified, and they all possess a large regulatory domain that covers about 60% of the protein. This regulation is accomplished by interaction with small molecules, posttranslational modifications, and mostly protein-protein interactions. In our search for new binding partners of the IP(3)R, we found that 90-kDa heat-shock protein (Hsp90) binds to the IP(3)R. This interaction increased on stimulation of HEK293T6.11 cells with insulin but not with G(q) protein-coupled receptor (G(q)PCR) agonists. Moreover, the Hsp90 inhibitor geldanamycin (GA) disrupted the interaction between Hsp90 and the IP(3)R. Pretreatment of HEK293T6.11 cells with GA greatly increased the intracellular Ca(2+) release induced by a G(q)PCR agonist. Insulin alone did not induce any intracellular Ca(2+) release. However, insulin diminished the intracellular Ca(2+) release induced by a G(q)PCR agonist. Interestingly, GA abolished the inhibitory effect of insulin on G(q)PCR-induced intracellular Ca(2+) release. Furthermore, in our search for a mechanistic explanation to this phenomenon, we found that inhibition of kinases activated downstream of the insulin receptor greatly increased the interaction between Hsp90 and the IP(3)R. Of greater interest, we found that the simultaneous inhibition of mammalian target of rapamycin and the Src kinase almost completely disrupted the interaction between Hsp90 and the IP(3)R. These results demonstrate that insulin promotes the interaction of Hsp90 with the IP(3)R to dampen its Ca(2+) release activity by a complex mechanism involving mammalian target of rapamycin and the Src kinase.


Subject(s)
Calcium/metabolism , HSP90 Heat-Shock Proteins/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Insulin/pharmacology , Benzoquinones/pharmacology , CSK Tyrosine-Protein Kinase , Calcium Signaling/drug effects , Carbachol/pharmacology , Cells, Cultured , Down-Regulation/drug effects , Humans , Lactams, Macrocyclic/pharmacology , Protein Binding/drug effects , Protein Kinases/metabolism , Protein Kinases/physiology , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/physiology , TOR Serine-Threonine Kinases , src-Family Kinases
16.
Mol Immunol ; 46(6): 1256-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19007993

ABSTRACT

The recessive lyp allele, which harbors a defective gimap5 (GTPase of immunity-associated nucleotide binding protein 5) gene, causes spontaneous apoptosis of T lymphocytes in the biobreeding diabetes-prone strain of rats. Mechanisms underlying the pro-survival function of GIMAP5 remain unclear. In this study, we show that gimap5(lyp/lyp) T cells display diminished calcium flux in response to thapsigargin or signaling via the T cell antigen receptor. This defect is manifested in mature single positive thymocytes, where the survival defect first occurs. We also show that GIMAP5 deficiency does not affect the thapsigargin-induced calcium release from the intracellular stores but impairs subsequent calcium entry across the plasma membrane. Our findings suggest that GIMAP5 is an important regulator of calcium response in T lymphocytes and impaired calcium signaling might underlie spontaneous apoptosis of gimap5(lyp/lyp) T cells.


Subject(s)
Calcium Signaling/immunology , GTP-Binding Proteins/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Animals , Animals, Genetically Modified , Calcium Signaling/physiology , Cell Membrane/physiology , GTP-Binding Proteins/genetics , In Vitro Techniques , Rats , T-Lymphocytes/drug effects , Thapsigargin/pharmacology , Thymus Gland/cytology
17.
Cell Calcium ; 45(3): 251-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19070363

ABSTRACT

Transient receptor potential canonical (TRPC) channels function as cation channels. In a previous study, we identified the molecular determinants involved in promoting TRPC subunit assembly. In the present study, we used size-exclusion chromatography assays to show that the N-terminus of TRPC4 can self-associate and form a tetramer in cellulo. We further showed that the N-terminus of TRPC4 self-associates via the ankyrin repeat domain and the region downstream from the coiled-coil domain. GST pull-down, yeast two-hybrid, and circular dichroism approaches demonstrated that both domains can self-associate. These findings indicated that the self-association of two distinct domains in the N-terminus of TRPC4 is involved in the assembly of the tetrameric channel.


Subject(s)
Protein Multimerization , TRPC Cation Channels/chemistry , TRPC Cation Channels/metabolism , Animals , Ankyrin Repeat , Humans , Mice , Peptide Fragments , Protein Structure, Tertiary , Structure-Activity Relationship , Two-Hybrid System Techniques
18.
Cell Physiol Biochem ; 22(1-4): 205-14, 2008.
Article in English | MEDLINE | ID: mdl-18769047

ABSTRACT

BACKGROUND/AIMS: The Alzheimer drug memantine (1-amino-3,5-dimethyl-adamantane) blocks the pore channel of the NMDA receptor. Since memantine also blocks the 5-HT(3) receptor, neuronal nicotinic receptor, and voltage-activated Na(+) channels, the purpose of our study was to verify whether memantine could influence other types of channels involved in the regulation of Ca(2+). METHODS: Free intracellular Ca(2+) concentrations in whole cells and in saponin-permeabilized cells were monitored spectrofluorometrically in HEK-293 cells stably expressing TRPC6. RESULTS: Memantine decreased the basal level of intracellular Ca(2+), increased the content of the intracellular Ca(2+) store, which in turn increased the agonist-induced intracellular Ca(2+) release, and increased the store-operated Ca(2+) entry. CONCLUSION: In addition to blocking the NMDA receptor, memantine also decreases the basal level of intracellular Ca(2+) and increases the sensitivity of cells to extracellular stimuli. All these effects may be of benefit in the treatment of Alzheimer's disease.


Subject(s)
Calcium Signaling/drug effects , Memantine/pharmacology , Animals , Calcium/metabolism , Carbachol/pharmacology , Cell Line , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Mice , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
19.
Am J Physiol Heart Circ Physiol ; 295(1): H21-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18502908

ABSTRACT

Extracellular purines and pyrimidines have major effects on cardiac rhythm and contraction. ATP/UTP are released during various physiopathological conditions, such as ischemia, and despite degradation by ectonucleotidases, their interstitial concentrations can markedly increase, a fact that is clearly associated with arrhythmia. In the present whole cell patch-clamp analysis on ventricular cardiomyocytes isolated from various mammalian species, ATP and UTP elicited a sustained, nonselective cationic current, I(ATP). UDP was ineffective, whereas 2'(3')-O-(4-benzoylbenzoyl)-ATP was active, suggesting that P2Y(2) receptors are involved. I(ATP) resulted from the binding of ATP(4-) to P2Y(2) purinoceptors. I(ATP) was maintained after ATP removal in the presence of guanosine 5'-[gamma-thio]triphosphate and was inhibited by U-73122, a PLC inhibitor. Single-channel openings are rather infrequent under basal conditions. ATP markedly increased opening probability, an effect prevented by U-73122. Two main conductance levels of 14 and 23 pS were easily distinguished. Similarly, in fura-2-loaded cardiomyocytes, Mn(2+) quenching and Ba(2+) influx were significant only in the presence of ATP or UTP. Adult rat ventricular cardiomyocytes expressed transient receptor potential channel TRPC1, -3, -4, and -7 mRNA and the TRPC3 and TRPC7 proteins that coimmunoprecipitated. Finally, the anti-TRPC3 antibody added to the patch pipette solution inhibited I(ATP). In conclusion, activation of P2Y(2) receptors, via a G protein and stimulation of PLCbeta, induces the opening of heteromeric TRPC3/7 channels, leading to a sustained, nonspecific cationic current. Such a depolarizing current could induce cell automaticity and trigger the arrhythmic events during an early infarct when ATP/UTP release occurs. These results emphasize a new, potentially deleterious role of TRPC channel activation.


Subject(s)
Adenosine Triphosphate/metabolism , Myocytes, Cardiac/metabolism , Receptors, Purinergic P2/metabolism , Signal Transduction , TRPC Cation Channels/metabolism , Uridine Triphosphate/metabolism , Animals , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/metabolism , Cell Membrane Permeability , Disease Models, Animal , Dogs , Estrenes/pharmacology , Humans , Male , Membrane Potentials , Mice , Mice, Knockout , Myocardial Infarction/complications , Myocardial Infarction/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Patch-Clamp Techniques , Phosphodiesterase Inhibitors/pharmacology , Phospholipase C beta/antagonists & inhibitors , Phospholipase C beta/metabolism , Pyrrolidinones/pharmacology , Rats , Rats, Wistar , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X , Receptors, Purinergic P2X4 , Receptors, Purinergic P2Y2 , Signal Transduction/drug effects
20.
Cell Calcium ; 43(5): 432-43, 2008 May.
Article in English | MEDLINE | ID: mdl-17850865

ABSTRACT

TRPCs function as cation channels in non-excitable cells. The N-terminal tails of all TRPCs contain an ankyrin-like repeat domain, one of the most common protein-protein interaction motifs. Using a yeast two-hybrid screening approach, we found that RNF24, a new membrane RING-H2 protein, interacted with the ankyrin-like repeat domain of TRPC6. GST pull-down and co-immunoprecipitation assays showed that RNF24 interacted with all TRPCs. Cell surface-labelling assays showed that the expression of TRPC6 at the surface of HEK 293T cells was greatly reduced when it was transiently co-transfected with RNF24. Confocal microscopy showed that TRPC3 and TRPC6 co-localized with RNF24 in a perinuclear compartment and that RNF24 co-localized with mannosidase II, a marker of the Golgi cisternae. Using a pulse-chase approach, we showed that RNF24 did not alter the maturation process of TRPC6. Moreover, in HEK 293T cells, RNF24 did not alter carbachol-induced Ca(2+) entry via endogenous channels or TRPC6. These results indicate that RNF24 interacts with TRPCs in the Golgi apparatus and affects TRPC intracellular trafficking without affecting their activity.


Subject(s)
Carrier Proteins/physiology , Membrane Proteins/physiology , TRPC Cation Channels/metabolism , Amino Acid Sequence , Ankyrin Repeat , Carbachol/pharmacology , Carrier Proteins/analysis , Carrier Proteins/chemistry , Cell Line , Cell Membrane/metabolism , Golgi Apparatus/chemistry , Humans , Membrane Proteins/analysis , Membrane Proteins/chemistry , Molecular Sequence Data , TRPC Cation Channels/chemistry , TRPC6 Cation Channel , Ubiquitin-Protein Ligases
SELECTION OF CITATIONS
SEARCH DETAIL
...