Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biochim Biophys Acta ; 1833(6): 1294-303, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23458835

RESUMO

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.


Assuntos
Canais de Cálcio Tipo T/metabolismo , Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Glicoproteínas de Membrana/metabolismo , Contração Muscular , Miócitos Cardíacos/metabolismo , Taquicardia , Animais , Western Blotting , Canais de Cálcio , Células Cultivadas , Eletrofisiologia , Imunoprecipitação , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/genética , Camundongos , Miócitos Cardíacos/citologia , Molécula 1 de Interação Estromal
2.
Biochem J ; 449(2): 353-64, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23098229

RESUMO

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.


Assuntos
Cálcio/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Mitocôndrias/metabolismo , Linfócitos T/metabolismo , Animais , Western Blotting , Membrana Celular/metabolismo , Células Cultivadas , Citoesqueleto/metabolismo , Proteínas de Ligação ao GTP/genética , Células HEK293 , Humanos , Transporte de Íons , Microscopia Confocal , Mutação , Ratos , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/citologia , Tubulina (Proteína)/metabolismo
3.
Proc Natl Acad Sci U S A ; 108(4): 1693-8, 2011 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-21220323

RESUMO

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.


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Glucose/farmacologia , Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Animais , Western Blotting , Encéfalo/metabolismo , Feminino , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Expressão Gênica , Teste de Tolerância a Glucose , Imuno-Histoquímica , Técnicas In Vitro , Insulina/genética , Secreção de Insulina , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
J Biol Chem ; 287(21): 17672-17681, 2012 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-22493444

RESUMO

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.


Assuntos
Sinalização do Cálcio/fisiologia , Proteínas Musculares/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Canais de Cátion TRPC/metabolismo , Androstadienos/farmacologia , Animais , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Carbacol/farmacologia , Cardiotônicos/farmacologia , Membrana Celular/genética , Membrana Celular/metabolismo , Cromonas/farmacologia , Células HEK293 , Humanos , Camundongos , Morfolinas/farmacologia , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/genética , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/genética , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Canais de Cátion TRPC/genética , Canal de Cátion TRPC6 , Wortmanina
5.
Biochim Biophys Acta ; 1808(12): 2807-18, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21864503

RESUMO

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.


Assuntos
Canais de Cátion TRPC/metabolismo , Zinco/metabolismo , Linhagem Celular , Colorimetria , Corantes Fluorescentes/metabolismo , Homeostase , Humanos , Canal de Cátion TRPC6
6.
J Biol Chem ; 285(52): 40534-43, 2010 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-20961851

RESUMO

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).


Assuntos
Miócitos de Músculo Liso/metabolismo , Proteína Quinase C-delta/metabolismo , Canais de Cátion TRPC/metabolismo , Substituição de Aminoácidos , Carbacol/farmacologia , Carcinógenos/farmacologia , Proliferação de Células/efeitos dos fármacos , Glomerulosclerose Segmentar e Focal/genética , Glomerulosclerose Segmentar e Focal/metabolismo , Células HEK293 , Humanos , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Isoenzimas/metabolismo , Mióticos/farmacologia , Mutação de Sentido Incorreto , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Proteína Quinase C-delta/antagonistas & inibidores , Proteína Quinase C-delta/genética , Serina/genética , Serina/metabolismo , Canais de Cátion TRPC/genética , Canal de Cátion TRPC6 , Acetato de Tetradecanoilforbol/farmacologia , Vasoconstritores/farmacologia , Vasopressinas/farmacologia
7.
Biochim Biophys Acta ; 1803(7): 805-12, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20346379

RESUMO

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.


Assuntos
Canais de Cátion TRPC/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Transporte Biológico/fisiologia , Membrana Celular/metabolismo , Células HeLa , Humanos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Canais de Cátion TRPC/genética , Canal de Cátion TRPC6 , Proteínas rab de Ligação ao GTP/genética
8.
J Immunol ; 183(4): 2747-57, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19620302

RESUMO

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.


Assuntos
Sinalização do Cálcio , Cavéolas/metabolismo , Caveolina 1/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator de Ativação de Plaquetas/fisiologia , Animais , Sítios de Ligação/imunologia , Células CHO , Sinalização do Cálcio/imunologia , Cavéolas/enzimologia , Cavéolas/imunologia , Linhagem Celular , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Ativação Enzimática/imunologia , Humanos , Microdomínios da Membrana/imunologia , Microdomínios da Membrana/metabolismo , Fragmentos de Peptídeos/metabolismo , Glicoproteínas da Membrana de Plaquetas/metabolismo , Glicoproteínas da Membrana de Plaquetas/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/fisiologia
9.
J Cell Physiol ; 225(2): 444-53, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20458742

RESUMO

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.


Assuntos
Cálcio/metabolismo , Receptor Muscarínico M1/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Transdução de Sinais/fisiologia , Canais de Cátion TRPC/metabolismo , Linhagem Celular , Membrana Celular/fisiologia , Retículo Endoplasmático/metabolismo , Regulação da Expressão Gênica/fisiologia , Humanos , Permeabilidade , Receptor Muscarínico M1/genética , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Canais de Cátion TRPC/genética
10.
Biochim Biophys Acta Mol Cell Res ; 1866(8): 1249-1259, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30951783

RESUMO

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.


Assuntos
Sinalização do Cálcio , Cálcio/metabolismo , Cardiomiopatia Dilatada/metabolismo , Proteínas Musculares/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Substituição de Aminoácidos , Animais , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/patologia , Células Cultivadas , Humanos , Proteínas Musculares/genética , Mutação de Sentido Incorreto , Miócitos Cardíacos/patologia , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Domínios Proteicos , Ratos , Ratos Sprague-Dawley , Molécula 1 de Interação Estromal/genética , Molécula 1 de Interação Estromal/metabolismo , Proteínas de Transporte Vesicular/genética
11.
Cell Calcium ; 43(5): 432-43, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-17850865

RESUMO

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.


Assuntos
Proteínas de Transporte/fisiologia , Proteínas de Membrana/fisiologia , Canais de Cátion TRPC/metabolismo , Sequência de Aminoácidos , Repetição de Anquirina , Carbacol/farmacologia , Proteínas de Transporte/análise , Proteínas de Transporte/química , Linhagem Celular , Membrana Celular/metabolismo , Complexo de Golgi/química , Humanos , Proteínas de Membrana/análise , Proteínas de Membrana/química , Dados de Sequência Molecular , Canais de Cátion TRPC/química , Canal de Cátion TRPC6 , Ubiquitina-Proteína Ligases
12.
Neuron ; 41(1): 153-63, 2004 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-14715142

RESUMO

Stimulation of adenylyl cyclase in the hippocampus is critical for memory formation. However, generation of cAMP signals within an optimal range for memory may require a balance between stimulatory and inhibitory mechanisms. The role of adenylyl cyclase inhibitory mechanisms for memory has not been addressed. One of the mechanisms for inhibition of adenylyl cyclase is through activation of G(i)-coupled receptors, a mechanism that could serve as a constraint on memory formation. Here we report that ablation of G(ialpha1) by gene disruption increases hippocampal adenylyl cyclase activity and enhances LTP in area CA1. Furthermore, gene ablation of G(ialpha1) or antisense oligonucleotide-mediated depletion of G(ialpha1) disrupted hippocampus-dependent memory. We conclude that G(ialpha1) provides a critical mechanism for tonic inhibition of adenylyl cyclase activity in the hippocampus. We hypothesize that loss of G(ialpha1) amplifies the responsiveness of CA1 postsynaptic neurons to stimuli that strengthen synaptic efficacy, thereby diminishing synapse-specific plasticity required for new memory formation.


Assuntos
Adenilil Ciclases/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Hipocampo/enzimologia , Potenciação de Longa Duração/fisiologia , Memória/fisiologia , Animais , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/antagonistas & inibidores , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Deleção de Genes , Hipocampo/fisiologia , Memória/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Toxina Pertussis/farmacologia
13.
Cell Physiol Biochem ; 22(1-4): 205-14, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18769047

RESUMO

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.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Memantina/farmacologia , Animais , Cálcio/metabolismo , Carbacol/farmacologia , Linhagem Celular , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Humanos , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Camundongos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo
14.
Cell Calcium ; 42(2): 225-32, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17368756

RESUMO

Thirteen years ago, it was suggested that exocytotic insertion of store-operated channels into the plasma membrane lead to increased Ca(2+) entry in non-excitable cells upon G protein-coupled or tyrosine kinase receptor stimulation. Since the discovery of the TRP channel superfamily and their involvement in receptor-induced Ca(2+) entry, many studies have shown that different members of the TRP superfamily translocate into the plasma membrane upon stimulation. While the exact molecular mechanism by which TRP channels insert into the plasma membrane is unknown, TRP-binding proteins have been shown to directly regulate this trafficking. This review summarizes recent advances related to the mechanism of TRP channel trafficking, focusing on the role of TRP-binding proteins.


Assuntos
Canais de Cálcio/metabolismo , Canais de Cátion TRPC/metabolismo , Animais , Proteínas de Transporte/metabolismo , Hormônios/farmacologia , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/efeitos da radiação , Proteínas SNARE/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo
15.
Biochim Biophys Acta ; 1763(10): 1051-8, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16962188

RESUMO

Heterotrimeric Gi proteins play a role in lipopolysaccharide (LPS) and Staphylococcus aureus (SA) activated signaling leading to inflammatory mediator production. We hypothesized that genetic deletion of Gi proteins would alter cytokine and chemokine production induced by LPS and SA. LPS- and heat killed SA-induced cytokine and chemokine production in splenocytes from wild type (WT), Galpha(i2) (-/-) or Galpha(i1/3) (-/-) mice were investigated. LPS- or SA-induced production of TNFalpha, IL-6, IFNgamma, IL-12, IL-17, GM-CSF, MIP-1alpha, MCP-1, MIG and IP-10 were significantly increased (1.2 to 33 fold, p<0.05) in splenocytes harvested from Galpha(i2)(-/-) mice compared with WT mice. The effect of Galpha(i) protein depletion was remarkably isoform specific. In splenocytes from Galpha(i1/3) (-/-) mice relative to WT mice, SA-induced IL-6, IFNgamma, GM-CSF, and IP-10 levels were decreased (59% to 86%, p<0.05), whereas other LPS- or SA-stimulated cytokines and chemokines were not different relative to WT mice. LPS- and SA-induced production of KC were unchanged in both groups of the genetic deficient mice. Splenocytes from both Galpha(i2) (-/-) and Galpha(i1/3) (-/-) mice did not exhibit changes in TLR2 and TLR4 expression. Also analysis of splenic cellular composition by flow cytometry demonstrated an increase in splenic macrophages and reduced CD4 T cells in both Galpha(i2) (-/-) and Galpha(i1/3) (-/-) mice relative to WT mice. The disparate response of splenocytes from the Galpha(i2) (-/-) relative to Galpha(i1/3) (-/-) mice therefore cannot be attributed to major differences in spleen cellular composition. These data demonstrate that G(i2) and G(i1/3) proteins are both involved and differentially regulate splenocyte inflammatory cytokine and chemokine production in a highly Gi isoform specific manner in response to LPS and Gram-positive microbial stimuli.


Assuntos
Quimiocinas/biossíntese , Citocinas/biossíntese , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/fisiologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Lipopolissacarídeos/farmacologia , Baço/metabolismo , Staphylococcus aureus/fisiologia , Animais , Células Cultivadas , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Regulação da Expressão Gênica , Camundongos , Camundongos Knockout , Isoformas de Proteínas , Sensibilidade e Especificidade , Baço/citologia , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo
16.
Biochimie ; 89(8): 926-37, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17532549

RESUMO

We synthesized a diacylglycerol (DAG)-containing arachidonic acid, i.e., 1-stearoyl-2-arachidonyl-sn-glycerol (SAG), and studied its implication in the modulation of canonical transient receptor potential sub-type 6 (TRPC6) channels in stably-transfected HEK-293 cells. SAG induced the influx of Ca(2+), and also of other bivalent cations like Ba(2+) and Sr(2+), in these cells. SAG-evoked Ca(2+) influx was not due to its metabolites as inhibitors of DAG-lipase (RHC80267) and DAG-kinase (R50922) failed to inhibit the response of the same. To emphasise that SAG exerts its action via its DAG configuration, but not due to the presence of stearic acid at sn-1 position, we synthesized 1-palmitoyl-2-arachidonyl-sn-glycerol (PAG). PAG-induced increases in [Ca(2+)](i) were not significantly different from those induced by SAG. For the comparative studies, we also synthesized the DAG-containing docosahexaenoic acid, i.e., 1-stearoyl-2-docosahexaenoyl-sn-glycerol (SDG). We observed that SDG and 1,2-dioctanoyl-sn-glycerol (DOG), a DAG analogue, also evoked increases in [Ca(2+)](i), which were lesser than those evoked by SAG. However, activation of TRPC6 channels by all the DAG molecular species (SAG, DOG and SDG) required Src kinases as the tyrosine kinase inhibitors, PP2 and SU6656, significantly attenuated the increases in [Ca(2+)](i) evoked by these agents. Moreover, disruption of lipid rafts with methyl-beta-cyclodextrin completely abolished SAG-, DOG- and SDG-induced increases in [Ca(2+)](i). The present study shows that SAG as well as SDG and DOG stimulate Ca(2+) influx through the activation of TRPC6 calcium channels which are regulated by Src kinases and intact lipid raft domains.


Assuntos
Diglicerídeos/farmacologia , Ácidos Docosa-Hexaenoicos/farmacologia , Canais de Cátion TRPC/metabolismo , Animais , Cálcio/metabolismo , Células Cultivadas , Diglicerídeos/síntese química , Ácidos Docosa-Hexaenoicos/síntese química , Relação Dose-Resposta a Droga , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Camundongos , Canal de Cátion TRPC6 , Quinases da Família src/metabolismo
17.
Front Immunol ; 8: 94, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28223986

RESUMO

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.

18.
Cell Signal ; 17(4): 437-45, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15601622

RESUMO

Mutations in the presenilin (PS) genes are linked to the development of early-onset Alzheimer's disease by a gain-of-function mechanism that alters proteolytic processing of the amyloid precursor protein (APP). Recent work indicates that Alzheimer's-disease-linked mutations in presenilin1 and presenilin2 attenuate calcium entry and augment calcium release from the endoplasmic reticulum (ER) in different cell types. However, the regulatory mechanisms underlying the altered profile of Ca(2+) signaling are unknown. The present study investigated the influence of two familial Alzheimer's-disease-linked presenilin2 variants (N141I and M239V) and a loss-of-function presenilin2 mutant (D263A) on the activity of the transient receptor potential canonical (TRPC)6 Ca(2+) entry channel. We show that transient coexpression of Alzheimer's-disease-linked presenilin2 mutants and TRPC6 in human embryonic kidney (HEK) 293T cells abolished agonist-induced TRPC6 activation without affecting agonist-induced endogenous Ca(2+) entry. The inhibitory effect of presenilin2 and the Alzheimer's-disease-linked presenilin2 variants was not due to an increase in amyloid beta-peptides in the medium. Despite the strong negative effect of the presenilin2 and Alzheimer's-disease-linked presenilin2 variants on agonist-induced TRPC6 activation, conformational coupling between inositol 1,4,5-trisphosphate receptor type 3 (IP(3)R3) and TRPC6 was unaffected. In cells coexpressing presenilin2 or the FAD-linked presenilin2 variants, Ca(2+) entry through TRPC6 could still be induced by direct activation of TRPC6 with 1-oleoyl-2-acetyl-sn-glycerol (OAG). Furthermore, transient coexpression of a loss-of-function PS2 mutant and TRPC6 in HEK293T cells enhanced angiotensin II (AngII)- and OAG-induced Ca(2+) entry. These results clearly indicate that presenilin2 influences TRPC6-mediated Ca(2+) entry into HEK293 cells.


Assuntos
Doença de Alzheimer/metabolismo , Cálcio/metabolismo , Canais Iônicos/metabolismo , Proteínas de Membrana/metabolismo , Doença de Alzheimer/genética , Secretases da Proteína Precursora do Amiloide , Ácido Aspártico Endopeptidases , Linhagem Celular , Endopeptidases/genética , Humanos , Proteínas de Membrana/genética , Mutação , Presenilina-2 , Canais de Cátion TRPC , Regulação para Cima
19.
Cell Calcium ; 37(2): 97-104, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15589990

RESUMO

In non-excitable cells, the inositol 1,4,5-trisphosphate receptor (IP3R) is an intracellular Ca2+ channel playing a major role in Ca2+ signaling. Three isoforms of IP3R have been identified and most cell types express different proportions of each isoform. The DT40 B lymphocyte cell line lacking all three IP3R isoforms (DT40IP3R-KO cells) represents an excellent model to re-express any recombinant IP3R and analyze its specific properties. In the study presented here, we confirmed that DT40IP3R-KO cells do not express any IP3-sensitive Ca2+ release channel. However, with an immunoblot approach and a [3H]IP3 binding approach we demonstrated the presence of a C-terminally truncated form of IP3R type III in the cytosolic fraction of DT40IP3R-KO cells. We further showed that this truncated IP3R retained the ability to couple to the Ca2+ entry channel TRPC6. Therefore, a word of caution is offered about the interpretation of results obtained in using DT40IP3R-KO cells to study the cellular mechanisms of Ca2+ entry.


Assuntos
Canais de Cálcio/genética , Inositol 1,4,5-Trifosfato/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Animais , Cálcio/metabolismo , Canais de Cálcio/biossíntese , Canais de Cálcio/imunologia , Células Cultivadas , Galinhas/genética , Galinhas/metabolismo , Immunoblotting , Receptores de Inositol 1,4,5-Trifosfato , Canais Iônicos/metabolismo , Receptores Citoplasmáticos e Nucleares/biossíntese , Receptores Citoplasmáticos e Nucleares/imunologia , Canais de Cátion TRPC , Trítio/metabolismo
20.
Br J Pharmacol ; 145(4): 405-14, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15806115

RESUMO

1 2-aminoethoxydiphenyl borate (2-APB) has been widely used to examine the roles of inositol 1,4,5-trisphosphate receptors (IP3Rs) and store-operated Ca2+ entry and is an emerging modulator of cationic channels encoded by transient receptor potential (TRP) genes. 2 Using Ca2+-indicator dye and patch-clamp recording we first examined the blocking effect of 2-APB on human TRPC5 channels expressed in HEK-293 cells. 3 The concentration-response curve has an IC50 of 20 microM and slope close to 1.0, suggesting one 2-APB molecule binds per channel. The blocking effect is not shared by other Ca2+ channel blockers including methoxyverapamil, nifedipine, N-propargylnitrendipine, or berberine. 4 In whole-cell and excised membrane patch recordings, 2-APB acts from the extracellular but not intracellular face of the membrane. 5 Block of TRPC5 by 2-APB is less at positive voltages, suggesting that it enters the electric field or acts by modulating channel gating. 6 2-APB also blocks TRPC6 and TRPM3 expressed in HEK-293 cells, but not TRPM2. 7 Block of TRP channels by 2-APB may be relevant to cell proliferation because 2-APB has a greater inhibitory effect on proliferation in cells overexpressing TRPC5. 8 Our data indicate a specific and functionally important binding site on TRPC5 that enables block by 2-APB. The site is only available via an extracellular route and the block shows mild voltage-dependence.


Assuntos
Compostos de Boro/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/fisiologia , Proteínas de Transporte de Cátions/fisiologia , Animais , Berberina/farmacologia , Canais de Cálcio/genética , Proteínas de Transporte de Cátions/genética , Linhagem Celular , Relação Dose-Resposta a Droga , Estimulação Elétrica , Galopamil/farmacologia , Humanos , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Nifedipino/farmacologia , Técnicas de Patch-Clamp , Canais de Cátion TRPC , Canal de Cátion TRPC6 , Canais de Cátion TRPM , Fatores de Tempo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA