Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Biochim Biophys Acta Mol Cell Res ; 1865(3): 463-469, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29223474

RESUMEN

Store-operated Ca2+ entry (SOCE) is a functionally relevant mechanism for Ca2+ influx present in electrically excitable and non-excitable cells. Regulation of Ca2+ entry through store-operated channels is essential to maintain an appropriate intracellular Ca2+ homeostasis and prevent cell damage. Calcium-release activated channels exhibit Ca2+-dependent inactivation mediated by two temporally separated mechanisms: fast Ca2+-dependent inactivation takes effect in the order of milliseconds and involves the interaction of Ca2+ with residues in the channel pore while slow Ca2+-dependent inactivation (SCDI) develops over tens of seconds, requires a global rise in [Ca2+]cyt and is a mechanism regulated by mitochondria. Recent studies have provided evidence that the protein SARAF (SOCE-associated regulatory factor) is involved in the mechanism underlying SCDI of Orai1. SARAF is an endoplasmic reticulum (ER) membrane protein that associates with STIM1 and translocate to plasma membrane-ER junctions in a STIM1-dependent manner upon store depletion to modulate SOCE. SCDI mediated by SARAF depends on the location of the STIM1-Orai1 complex within a PI(4,5)P2-rich microdomain. SARAF also interacts with Orai1 and TRPC1 in cells endogenously expressing STIM1 and cells with a low STIM1 expression and modulates channel function. This review focuses on the modulation by SARAF of SOCE and other forms of Ca2+ influx mediated by Orai1 and TRPC1 in order to provide spatio-temporally regulated Ca2+ signals.


Asunto(s)
Señalización del Calcio/genética , Calcio/metabolismo , Retículo Endoplásmico/genética , Proteínas de la Membrana/genética , Canales de Calcio/genética , Membrana Celular/genética , Humanos , Proteínas Sensoras del Calcio Intracelular , Mitocondrias/genética , Proteína ORAI1/genética , Molécula de Interacción Estromal 1/genética , Canales Catiónicos TRPC/genética
2.
Adv Exp Med Biol ; 898: 67-85, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27161225

RESUMEN

Na(+)/Ca(2+) exchangers (NCXs) have traditionally been viewed principally as a means of Ca(2+) removal from non-excitable cells. However there has recently been increasing interest in the operation of NCXs in reverse mode acting as a means of eliciting Ca(2+) entry into these cells. Reverse mode exchange requires a significant change in the normal resting transmembrane ion gradients and membrane potential, which has been suggested to occur principally via the coupling of NCXs to localised Na(+) entry through non-selective cation channels such as canonical transient receptor potential (TRPC) channels. Here we review evidence for functional or physical coupling of NCXs to non-selective cation channels, and how this affects NCX activity in non-excitable cells. In particular we focus on the potential role of nanojunctions, where the close apposition of plasma and intracellular membranes may help create the conditions needed for the generation of localised rises in Na(+) concentration that would be required to trigger reverse mode exchange.


Asunto(s)
Intercambiador de Sodio-Calcio/metabolismo , Canales Catiónicos TRPC/metabolismo , Calcio/metabolismo , Humanos , Transporte Iónico , Sodio/metabolismo
3.
Cell Calcium ; 101: 102522, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34968774

RESUMEN

Human platelets regulate agonist-evoked Ca2+ signalling through Ca2+ release from and sequestration into acidic organelles. Previous studies have pharmacologically characterised the presence of a Ca2+-H+ exchanger in these organelles. This exchanger appears to regulate a secondary plateau phase in agonist-evoked cytosolic Ca2+ signals in fura-2-loaded human platelets. Here we demonstrate that cytochalasin D treatment removes the secondary plateau in ADP-evoked Ca2+ signals elicited in the absence of external Ca2+. This effect was reversed by pretreatment with nigericin, a K+/H+ exchanger that short-circuits the Ca2+-H+ exchanger. Using Fluo-5N- or Lysosensor Green-loaded cells, cytochalasin D was found to enhance Ca2+ sequestration into acidic organelles by preventing their alkalinisation. Additional experiments demonstrated that ADP-evoked alkalinisation of acidic organelles and subsequent slowing of acidic organellar Ca2+ sequestration was mediated by autocrine 5-HT signalling. Enhancing this 5-HT signalling using fluoxetine overcame the inhibitory effect of cytochalasin D on ADP-evoked Ca2+ signals, indicating that cytochalasin D interferes with 5-HT autocrine signalling. The ability of Cytochalasin D to interfere with autocrine 5-HT signalling was downstream of the 5-HT2A receptor as secretion of [3H]-5-HT from ADP-stimulated human platelets was not reduced. These data provide the first evidence that the pH gradient across acidic organelles is dynamically regulated upon human platelet activation, and that this can play a significant role in controlling human platelet function by modulating Ca2+-H+ exchange and so [Ca2+]i.


Asunto(s)
Calcio , Serotonina , Citoesqueleto de Actina , Plaquetas , Humanos , Orgánulos
4.
J Biol Chem ; 284(49): 33750-62, 2009 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-19815551

RESUMEN

Platelets are activated by adhesion to vascular collagen via the immunoglobulin receptor, glycoprotein VI (GPVI). This causes potent signaling toward activation of phospholipase Cgamma2, which bears similarity to the signaling pathway evoked by T- and B-cell receptors. Phosphoinositide 3-kinase (PI3K) plays an important role in collagen-induced platelet activation, because this activity modulates the autocrine effects of secreted ADP. Here, we identified the PI3K isoforms directly downstream of GPVI in human and mouse platelets and determined their role in GPVI-dependent thrombus formation. The targeting of platelet PI3Kalpha or -beta strongly and selectively suppressed GPVI-induced Ca(2+) mobilization and inositol 1,4,5-triphosphate production, thus demonstrating enhancement of phospholipase Cgamma2 by PI3Kalpha/beta. That PI3Kalpha and -beta have a non-redundant function in GPVI-induced platelet activation and thrombus formation was concluded from measurements of: (i) serine phosphorylation of Akt, (ii) dense granule secretion, (iii) intracellular Ca(2+) increases and surface expression of phosphatidylserine under flow, and (iv) thrombus formation, under conditions where PI3Kalpha/beta was blocked or p85alpha was deficient. In contrast, GPVI-induced platelet activation was insensitive to inhibition or deficiency of PI3Kdelta or -gamma. Furthermore, PI3Kalpha/beta, but not PI3Kgamma, contributed to GPVI-induced Rap1b activation and, surprisingly, also to Rap1b-independent platelet activation via GPVI. Together, these findings demonstrate that both PI3Kalpha and -beta isoforms are required for full GPVI-dependent platelet Ca(2+) signaling and thrombus formation, partly independently of Rap1b. This provides a new mechanistic explanation for the anti-thrombotic effect of PI3K inhibition and makes PI3Kalpha an interesting new target for anti-platelet therapy.


Asunto(s)
Plaquetas/metabolismo , Fosfatidilinositol 3-Quinasas/química , Fosfatidilinositol 3-Quinasas/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Trombosis/patología , Animales , Calcio/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/genética , Activación Plaquetaria , Agregación Plaquetaria , Isoformas de Proteínas , Transducción de Señal , Trombosis/metabolismo
5.
Biochim Biophys Acta ; 1793(2): 223-30, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19061922

RESUMEN

Store-operated calcium entry (SOCE) is a major mechanism for Ca(2+) influx. Since SOCE was first proposed two decades ago many techniques have been used in attempting to identify the nature of store-operated Ca(2+) (SOC) channels. The first identified and best-characterised store-operated current is I(CRAC), but a number of other currents activated by Ca(2+) store depletion have also been described. TRPC proteins have long been proposed as SOC channel candidates; however, whether any of the TRPCs function as SOC channels remains controversial. This review attempts to provide an overview of the arguments in favour and against the role of TRPC proteins in the store-operated mechanisms of agonist-activated Ca(2+) entry.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Humanos , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Proteínas de la Membrana/metabolismo
6.
Platelets ; 21(6): 445-50, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20482247

RESUMEN

Platelet activation is critically regulated by an increase in intracellular calcium concentration ([Ca2+](i)). Although Ca2+ release from intracellular Ca2+ stores and subsequent store-operated Ca2+ entry are often thought to be the major contributors to increases in [Ca2+](i) evoked by most agonists, high concentrations of thrombin activate a Ca2+ entry pathway that is independent of Ca2+ store depletion (known as 'non-capacitative cation entry'-NCCE). The channel that conducts NCCE has not previously been clearly identified, and the mechanisms that regulate its activation are also unknown. Here we have investigated NCCE using fura-2-loaded human platelets. To investigate NCCE independently of other Ca2+ signaling pathways, the intracellular Ca2+ stores were first rapidly depleted in the absence of extracellular Ca2+. Sr2+ was then added to monitor maximal store-operated cation influx. Thrombin was then added to stimulate NCCE. Flufenamic acid, which inhibits Ca2+ entry through most TRPC isoforms, but potentiates entry through TRPC6, was found to block store-operated cation entry. In contrast, thrombin-induced NCCE was increased, suggesting the possible involvement of TRPC6. Since TRPC6 is regulated by Src family tyrosine kinases in some cells, we investigated the possible role of this kinase family in NCCE. PP2, a Src family tyrosine kinase inhibitor, completely abolished thrombin-induced NCCE. Furthermore, NCCE was enhanced by phenylarsine oxide and could be directly induced by vanadyl hydroperoxide, both tyrosine phosphatase inhibitors. These data indicate that Src family tyrosine kinase activation is a required step in NCCE activation. In conclusion NCCE may be an important regulator of platelet activation when local thrombin concentrations are high.


Asunto(s)
Plaquetas/metabolismo , Calcio/sangre , Trombina/metabolismo , Familia-src Quinasas/sangre , Plaquetas/enzimología , Canales de Calcio/sangre , Cationes Bivalentes/metabolismo , Activación Enzimática , Humanos , Activación Plaquetaria/efectos de los fármacos , Canales Catiónicos TRPC/metabolismo , Canal Catiónico TRPC6
7.
Cell Signal ; 20(2): 337-46, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18068335

RESUMEN

Two agonist-releasable Ca(2+)stores have been identified in human platelets differentiated by the distinct sensitivity of their SERCA isoforms to thapsigargin (TG) and 2,5-di-(tert-butyl)-1,4-hydroquinone (TBHQ). Here we have examined whether the SERCA isotypes might be involved in store-operated Ca(2+)entry (SOCE) activated by the physiological agonist thrombin in human platelets. Ca(2+)-influx evoked by thrombin (0.01 U/mL) reached a maximum after 3 min, which was consistent with the decrease in the Ca(2+)content in the stores; afterwards, the extent of SOCE decreased with no correlation with the accumulation of Ca(2+)in the stores. Inhibition of SERCA2b, by 10 nM TG, and SERCA3, with 20 microM TBHQ, individually or simultaneously, accelerated Ca(2+) store discharge and subsequently enhanced the extent of SOCE stimulated by thrombin. In addition, TG and TBHQ modified the time course of thrombin-evoked SOCE from a transient to a sustained increase in Ca(2+) influx, which reveals a negative role for SERCAs in the regulation of SOCE. This effect was consistent under conditions that inhibit Ca(2+) extrusion by PMCA or the Na(+)/Ca(2+) exchanger. Coimmunoprecipitation experiments revealed that thrombin stimulates direct interaction between SERCA2b and 3 with the hTRPC1 channel, an effect that was found to be independent of SERCA activity. In summary, our results suggest that SERCA2b and 3 modulate thrombin-stimulated SOCE probably by direct interaction with the hTRPC1 channel in human platelets.


Asunto(s)
Plaquetas/enzimología , Señalización del Calcio , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Plaquetas/efectos de los fármacos , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Humanos , Inmunoprecipitación , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Isoenzimas/metabolismo , Unión Proteica/efectos de los fármacos , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/antagonistas & inhibidores , Canales Catiónicos TRPC/metabolismo , Trombina/farmacología , Factores de Tiempo
9.
Int Rev Cytol ; 256: 35-67, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17241904

RESUMEN

The transient receptor potential (TRP) family of ion channels is composed of more than 50 functionally versatile cation-permeant ion channels expressed in most mammalian cell types. Considerable research has been brought to bear on the members of this family, especially with regard to their possible role as store-operated calcium channels, although studies have provided evidence that TRP channels exhibit a number of regulatory and functional aspects. Endogenous and transiently expressed TRP channels can be activated by different mechanisms grouped into four main categories: receptor-operated activation, store depletion-mediated activation, ligand-induced activation, and direct activation. This article reviews the biochemical characteristics of the different members of the TRP family and summarizes their involvement in a number of physiological events ranging from sensory transduction to development, which might help in understanding the relationship between TRP channel dysfunction and the development of several diseases.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/fisiología , Canales de Potencial de Receptor Transitorio/fisiología , Animales , Calcio/metabolismo , Señalización del Calcio , Humanos , Modelos Biológicos , Sistemas de Mensajero Secundario , Canales de Potencial de Receptor Transitorio/genética
10.
Cell Signal ; 19(10): 2147-54, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17681754

RESUMEN

Two mechanisms for store-operated Ca(2+) entry (SOCE) regulated by two independent Ca(2+) stores, the dense tubular system (DTS) and the acidic stores, have been described in platelets. We have previously suggested that coupling between the type II IP(3) receptor (IP(3)RII) and hTRPC1, involving reorganization of the actin microfilaments, play an important role in SOCE. However, the involvement of the tubulin microtubules, located beneath the plasma membrane, remains unclear. Here we show that the microtubule disrupting agent colchicine reduced Ca(2+) entry stimulated by low concentrations (0.1 U/mL) of thrombin, which activates SOCE mostly by depleting acidic Ca(2+)-store. Consistently, colchicine reduced SOCE activated by 2,5 di-(tertbutyl)-1,4-hydroquinone (TBHQ), which selectively depletes the acidic Ca(2+) stores. In contrast, colchicine enhanced SOCE mediated by depletion of the DTS, induced by high concentrations of thapsigargin (TG), which depletes both the acidic Ca(2+) stores and the DTS, the major releasable Ca(2+) store in platelets. These findings were confirmed by using Sr(2+) as a surrogate for Ca(2+) entry. Colchicine attenuated the coupling between IP(3)RII and hTRPC1 stimulated by thrombin while it enhanced that evoked by TG. Paclitaxel, which induces microtubular stabilization and polymerization, exerted the opposite effects on thrombin- and TG-evoked SOCE and coupling between IP(3)RII and hTRPC1 compared with colchicine. Neither colchicine nor paclitaxel altered the ability of platelets to extrude Ca(2+). These findings suggest that tubulin microtubules play a dual role in SOCE, acting as a barrier that prevents constitutive SOCE regulated by DTS, but also supporting SOCE mediated by the acidic Ca(2+) stores.


Asunto(s)
Plaquetas/metabolismo , Calcio/metabolismo , Microtúbulos/fisiología , Tubulina (Proteína)/fisiología , Citoesqueleto de Actina/fisiología , Plaquetas/efectos de los fármacos , Plaquetas/ultraestructura , Colchicina/farmacología , Humanos , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Transporte Iónico , Paclitaxel/farmacología , Canales Catiónicos TRPC/metabolismo , Tapsigargina/antagonistas & inhibidores , Trombina/antagonistas & inhibidores , Moduladores de Tubulina/farmacología
11.
Sci Rep ; 8(1): 16677, 2018 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-30420683

RESUMEN

Citalopram, a selective serotonin reuptake inhibitor (SSRI), inhibits platelet function in vitro. We have previously shown that this action is independent of citalopram's ability to block serotonin uptake by the serotonin transporter and must therefore be mediated via distinct pharmacological mechanisms. We now report evidence for two novel and putative mechanisms of citalopram-induced platelet inhibition. Firstly, in platelets, citalopram blocked U46619-induced Rap1 activation and subsequent platelet aggregation, but failed to inhibit U46619-induced increases in cytosolic Ca2+. Similarly, in neutrophils, citalopram inhibited Rap1 activation and downstream functions but failed to block PAF-induced Ca2+ mobilisation. In a cell-free system, citalopram also reduced CalDAG-GEFI-mediated nucleotide exchange on Rap1B. Secondly, the binding of anti-GPVI antibodies to resting platelets was inhibited by citalopram. Furthermore, citalopram-induced inhibition of GPVI-mediated platelet aggregation was instantaneous, reversible and displayed competitive characteristics, suggesting that these effects were not caused by a reduction in GPVI surface expression, but by simple competitive binding. In conclusion, we propose two novel, putative and distinct inhibitory mechanisms of action for citalopram: (1) inhibition of CalDAG-GEFI/Rap1 signalling, and (2) competitive antagonism of GPVI in platelets. These findings may aid in the development of novel inhibitors of CalDAG-GEFI/Rap1-dependent nucleotide exchange and novel GPVI antagonists.


Asunto(s)
Citalopram/farmacología , Neutrófilos/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Agregación Plaquetaria/efectos de los fármacos , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Calcio/metabolismo , Citosol/metabolismo , Humanos , L-Lactato Deshidrogenasa/metabolismo , Modelos Biológicos , Neutrófilos/citología , Glicoproteínas de Membrana Plaquetaria/metabolismo
12.
Sci Rep ; 8(1): 3494, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29472624

RESUMEN

Citalopram prevents serotonin (5-HT) uptake into platelets by blocking the serotonin reuptake transporter (SERT). Although some clinical data suggest that selective serotonin reuptake inhibitors (SSRIs) may affect haemostasis and thrombosis, these poorly-characterised effects are not well understood mechanistically and useful in vitro data is limited. We sought to determine whether the inhibitory effects of citalopram on platelets are mediated via its pharmacological inhibition of 5-HT transport. We quantified the inhibitory potency of (RS)-, (R)- and (S)-citalopram on platelet function. If SERT blockade is the primary mechanism for citalopram-mediated platelet inhibition, these potencies should show quantitative congruence with inhibition of 5-HT uptake. Our data show that citalopram inhibits platelet aggregation, adhesion and thromboxane production with no difference in potency between (R)- and (S)-isomers. By contrast, citalopram had a eudysmic ratio of approximately 17 (S > R) for SERT blockade. Furthermore, nanomolar concentrations of citalopram inhibited 5-HT uptake into platelets but had no effect on other platelet functions, which were inhibited by micromolar concentrations. Our data indicate that citalopram-induced inhibition of platelets in vitro is not mediated by blockade of 5-HT transport. This raises a new question for future investigation: by what mechanism(s) does citalopram inhibit platelets?


Asunto(s)
Citalopram/farmacología , Agregación Plaquetaria/efectos de los fármacos , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Serotonina/genética , Animales , Plaquetas/efectos de los fármacos , Plaquetas/patología , Voluntarios Sanos , Humanos , Ratones , Fosforilación , Agregación Plaquetaria/genética , Conejos , Serotonina/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Tromboxano A2/biosíntesis , Tromboxano A2/genética
13.
Cell Calcium ; 41(2): 169-78, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16884770

RESUMEN

Here, we report a novel role for the cysteine protease calpain in store-operated calcium entry. Several structurally and mechanistically unrelated inhibitors of calpain inhibited Ca2+ entry activated in human platelets by thapsigargin-evoked Ca2+ store depletion or the physiological agonist thrombin, whereas inhibitors of other cysteine proteases were without effect. The use of the cell-permeable fluorogenic calpain substrate 7-amino-4-chloromethylcoumarin, t-BOC-l-leucyl-l-methionine amide revealed rapid activation of calpain which was closely temporally correlated with Ca2+ store depletion even in the absence of a rise in cytosolic [Ca2+]. Calpain inhibition prevented the tyrosine phosphorylation of several proteins upon Ca2+ store depletion, suggesting that calpain may lie upstream of protein tyrosine phosphorylation that is known to be required for the activation of store-operated Ca2+ entry in human platelets. Earlier studies using calpain inhibitors may need reinterpretation in the light of this finding that calpain plays a role in the activation of physiological Ca2+ entry pathways.


Asunto(s)
Plaquetas/metabolismo , Señalización del Calcio , Calcio/metabolismo , Calpaína/fisiología , Plaquetas/enzimología , Calpaína/antagonistas & inhibidores , Células Cultivadas , Inhibidores de Cisteína Proteinasa/farmacología , Dipéptidos/farmacología , Interacciones Farmacológicas , Humanos , Fosforilación , Activación Plaquetaria/efectos de los fármacos , Estroncio/metabolismo , Tapsigargina/farmacología , Trombina/farmacología , Tirosina/metabolismo
14.
Cell Calcium ; 42(6): 606-17, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17383000

RESUMEN

We have previously demonstrated a role for the reorganization of the actin cytoskeleton in store-operated calcium entry (SOCE) in human platelets and interpreted this as evidence for a de novo conformational coupling step in SOCE activation involving the type II IP(3) receptor and the platelet hTRPC1-containing store-operated channel (SOC). Here, we present evidence challenging this model. The actin polymerization inhibitors cytochalasin D or latrunculin A significantly reduced Ca2+ but not Mn2+ or Na+ entry into thapsigargin (TG)-treated platelets. Jasplakinolide, which induces actin polymerization, also inhibited Ca2+ but not Mn2+ or Na+ entry. However, an anti-hTRPC1 antibody inhibited TG-evoked entry of all three cations, indicating that they all permeate an hTRPC1-containing store-operated channel (SOC). These results indicate that the reorganization of the actin cytoskeleton is not involved in SOC activation. The inhibitors of the Na+/Ca2+ exchanger (NCX), KB-R7943 or SN-6, caused a dose-dependent inhibition of Ca2+ but not Mn2+ or Na+ entry into TG-treated platelets. The effects of the NCX inhibitors were not additive with those of actin polymerization inhibitors, suggesting a common point of action. These results indicate a role for two Ca2+ permeable pathways activated following Ca2+ store depletion in human platelets: A Ca2+-permeable, hTRPC1-containing SOC and reverse Na+/Ca2+ exchange, which is activated following Na+ entry through the SOC and requires a functional actin cytoskeleton.


Asunto(s)
Actinas/metabolismo , Plaquetas/metabolismo , Calcio/metabolismo , Citoesqueleto/metabolismo , Intercambiador de Sodio-Calcio/fisiología , Compuestos de Bencilo/farmacología , Plaquetas/efectos de los fármacos , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Calcio/farmacología , Citocalasina D/farmacología , Depsipéptidos/farmacología , Humanos , Manganeso/metabolismo , Potasio/metabolismo , Potasio/farmacología , Sodio/metabolismo , Sodio/farmacología , Intercambiador de Sodio-Calcio/antagonistas & inhibidores , Tapsigargina/farmacología , Tiazolidinas/farmacología , Tiourea/análogos & derivados , Tiourea/farmacología
15.
FEBS Lett ; 580(22): 5313-20, 2006 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-16979629

RESUMEN

Platelet activation by potent, Ca(2+)-mobilizing agonists results in shedding of microparticles that are active in coagulation. Here we show that platelets under storage produce procoagulant microparticles in the absence of agonist. Microparticle formation by resting platelets results from alphaIIbbeta3 signaling to destabilization of the actin cytoskeleton in the absence of calpain activation. Integrin-mediated spreading of platelets over fibrinogen similarly results in microparticle formation. After transfusion of stored platelet preparations to thrombocytopenic patients, the microparticles contribute to coagulant activity in vivo.


Asunto(s)
Plaquetas/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Coagulantes/metabolismo , Activación Plaquetaria/efectos de los fármacos , Preservación Biológica , Transducción de Señal , Citoesqueleto de Actina/metabolismo , Coagulación Sanguínea/efectos de los fármacos , Transfusión de Componentes Sanguíneos/efectos adversos , Plaquetas/citología , Calpaína/metabolismo , Activación Enzimática/efectos de los fármacos , Humanos , Integrina beta3/metabolismo , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Trombocitopenia/metabolismo , Trombocitopenia/terapia
16.
Metallomics ; 8(1): 91-100, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26434726

RESUMEN

Following platelet adhesion and primary activation at sites of vascular injury, secondary platelet activation is induced by soluble platelet agonists, such as ADP, ATP, thrombin and thromboxane. Zinc ions are also released from platelets and damaged cells and have been shown to act as a platelet agonist. However, the mechanism of zinc-induced platelet activation is not well understood. Here we show that exogenous zinc gains access to the platelet cytosol and induces full platelet aggregation that is dependent on platelet protein tyrosine phosphorylation, PKC and integrin αIIbß3 activity and is mediated by granule release and secondary signalling. ZnSO4 increased the binding affinity of GpVI, but not integrin α2ß1. Low concentrations of ZnSO4 potentiated platelet aggregation by collagen-related peptide (CRP-XL), thrombin and adrenaline. Chelation of intracellular zinc reduced platelet aggregation induced by a number of different agonists, inhibited zinc-induced tyrosine phosphorylation and inhibited platelet activation in whole blood under physiologically relevant flow conditions. Our data are consistent with a transmembrane signalling role for zinc in platelet activation during thrombus formation.


Asunto(s)
Membrana Celular/metabolismo , Fosfotirosina/metabolismo , Activación Plaquetaria/efectos de los fármacos , Zinc/farmacología , Animales , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Proteínas Sanguíneas/metabolismo , Proteínas Portadoras/farmacología , Bovinos , Membrana Celular/efectos de los fármacos , Citosol/efectos de los fármacos , Citosol/metabolismo , Epinefrina/farmacología , Etilenodiaminas/farmacología , Humanos , Péptidos/farmacología , Fosforilación/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Glicoproteínas de Membrana Plaquetaria/metabolismo , Trombina/farmacología , Regulación hacia Arriba/efectos de los fármacos , Zinc/metabolismo
17.
Thromb Haemost ; 94(4): 839-45, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16270640

RESUMEN

We have previously suggested that the human homologue of the Drosophila transient receptor potential protein, TRPC1, is involved in conducting store-operated Ca2+ entry (SOCE) in human platelets since an antibody raised against the pore-forming region of TRPC1 inhibited SOCE. Here we have investigated plasma membrane expression of TRPC1 in human platelets and have probed for the presence of other TRPC proteins in these cells. Biotinylation revealed the presence of TRPC1 in the plasma membrane of resting platelets. Surface expression was not detectibly changed following Ca2+ store depletion or stimulation with thrombin. Western blotting demonstrated the presence of TRPC1, TRPC3, TRPC4, TRPC5 and TRPC6 in platelet lysates. TRPC1, TRPC4 and TRPC5 coimmunoprecipitated, as did TRPC3 and TRPC6. TRPC1, TRPC4 and TRPC5 were associated with detergent-resistant platelet membranes, from which they were partially released when the cells were cholesterol-depleted using methyl-beta-cyclodextrin. The distributions of TRPC3 and TRPC6 between soluble and membrane fractions were not affected by methyl-beta-cyclodextrin treatment. These results suggest that TRPC1, TRPC4 and TRPC5 form a heteromultimer associated with platelet lipid raft domains, whereas TRPC3 and TRPC6 associate independently of lipid rafts.


Asunto(s)
Plaquetas/metabolismo , Microdominios de Membrana/metabolismo , Canales Catiónicos TRPC/metabolismo , Biotinilación , Membrana Celular/metabolismo , Humanos , Transducción de Señal/fisiología , Canal Catiónico TRPC6
18.
Biochem J ; 375(Pt 3): 697-704, 2003 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-12908873

RESUMEN

Depletion of intracellular Ca2+ stores results in the activation of SMCE (store-mediated Ca2+ entry) in many cells. The mechanism of activation of SMCE is poorly understood. In human platelets, a secretion-like coupling model may be involved. This proposes that store depletion results in trafficking of portions of the endoplasmic reticulum to the plasma membrane, enabling coupling between proteins in the two membranes. In support of this, we have shown that, in human platelets, agonist-evoked Ca2+ store depletion results in de novo and reversible coupling of the Ins P3RII [type II inositol (1,4,5)trisphosphate receptor] with the putative Ca2+ entry channel hTRPC1 [human canonical transient receptor potential 1 (protein); Rosado, Brownlow and Sage (2002) J. Biol. Chem. 277, 42157-42163]. A crucial test of the hypothesis that this coupling activates SMCE is that it should occur rapidly enough to account for agonist-evoked Ca2+ entry. In the present study, we have used quenched- and stopped-flow approaches to determine the latencies of thrombin-evoked coupling of Ins P3RII with hTRPC1 and of thrombin-evoked bivalent cation entry using Mn2+ quenching of fura 2 fluorescence. Thrombin-evoked Mn2+ entry was detected with a latency of 0.81+/-0.07 s (S.E.M., n =7) or 1.36+/-0.09 s (S.E.M., n =7) at a concentration of 1.0 or 0.1 unit/ml respectively. Coupling between Ins P3RII and hTRPC1, assessed at 100 ms intervals, was first detected with a latency of 0.9 or 1.4 s after stimulation with thrombin at a concentration of 1.0 or 0.1 unit/ml respectively. These results support the hypothesis that de novo coupling of Ins P3RII with hTRPC1 could activate SMCE in human platelets.


Asunto(s)
Plaquetas/metabolismo , Canales de Calcio/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Plaquetas/efectos de los fármacos , Calcio/metabolismo , Cationes Bivalentes/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Humanos , Indoles/farmacología , Receptores de Inositol 1,4,5-Trifosfato , Manganeso/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/agonistas , Canales Catiónicos TRPC , Trombina/farmacología , Factores de Tiempo
19.
Trends Cardiovasc Med ; 12(5): 229-34, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12161078

RESUMEN

Store-mediated Ca(2+) entry (SMCE) is the main pathway for Ca(2+) influx in platelets and other nonexcitable cells, yet how depletion of the intracellular Ca(2+) stores leads to the activation of Ca(2+) entry across the plasma membrane remains unclear. Recent work in platelets favors a secretionlike conformational coupling mechanism involving the Ca(2+)-permeable channel protein, Trp1, in the plasma membrane and the type-II inositol 1,4,5-trisphosphate receptor in the membrane of the Ca(2+) store, which is located in the endoplasmic reticulum. Extracellular signal-regulated kinases (ERKs) are common participants in a broad variety of signal transduction pathways in human platelets, and inactivation of the ERK cascade has been shown to reduce Ca(2+) entry stimulated by thapsigargin or thrombin. The role of ERK in SMCE into human platelets was found to be independent of the cytoskeleton and a downstream effector of the small guanosine-triphosphate-binding protein Ras.


Asunto(s)
Plaquetas/fisiología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Activación Plaquetaria , 1-Fosfatidilinositol 4-Quinasa/metabolismo , Calcio/sangre , Calcio/metabolismo , Humanos , Proteína Quinasa 3 Activada por Mitógenos
20.
Cell Calcium ; 58(6): 577-88, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26434503

RESUMEN

Rises in cytosolic Ca(2+) concentration ([Ca(2+)]cyt) are central in platelet activation, yet many aspects of the underlying mechanisms are poorly understood. Most studies examine how experimental manipulations affect agonist-evoked rises in [Ca(2+)]cyt, but these only monitor the net effect of manipulations on the processes controlling [Ca(2+)]cyt (Ca(2+) buffering, sequestration, release, entry and removal), and cannot resolve the source of the Ca(2+) or the transporters or channels affected. To investigate the effects of protein kinase C (PKC) on platelet Ca(2+) signalling, we here monitor Ca(2+) flux around the platelet by measuring net Ca(2+) fluxes to or from the extracellular space and the intracellular Ca(2+) stores, which act as the major sources and sinks for Ca(2+) influx into and efflux from the cytosol, as well as monitoring the cytosolic Na(+) concentration ([Na(+)]cyt), which influences platelet Ca(2+) fluxes via Na(+)/Ca(2+) exchange. The intracellular store Ca(2+) concentration ([Ca(2+)]st) was monitored using Fluo-5N, the extracellular Ca(2+) concentration ([Ca(2+)]ext) was monitored using Fluo-4 whilst [Ca(2+)]cyt and [Na(+)]cyt were monitored using Fura-2 and SFBI, respectively. PKC inhibition using Ro-31-8220 or bisindolylmaleimide I potentiated ADP- and thrombin-evoked rises in [Ca(2+)]cyt in the absence of extracellular Ca(2+). PKC inhibition potentiated ADP-evoked but reduced thrombin-evoked intracellular Ca(2+) release and Ca(2+) removal into the extracellular medium. SERCA inhibition using thapsigargin and 2,5-di(tert-butyl) l,4-benzohydroquinone abolished the effect of PKC inhibitors on ADP-evoked changes in [Ca(2+)]cyt but only reduced the effect on thrombin-evoked responses. Thrombin evokes substantial rises in [Na(+)]cyt which would be expected to reduce Ca(2+) removal via the Na(+)/Ca(2+) exchanger (NCX). Thrombin-evoked rises in [Na(+)]cyt were potentiated by PKC inhibition, an effect which was not due to altered changes in non-selective cation permeability of the plasma membrane as assessed by Mn(2+) quench of Fura-2 fluorescence. PKC inhibition was without effect on thrombin-evoked rises in [Ca(2+)]cyt following SERCA inhibition and either removal of extracellular Na(+) or inhibition of Na(+)/K(+)-ATPase activity by removal of extracellular K(+) or treatment with digoxin. These data suggest that PKC limits ADP-evoked rises in [Ca(2+)]cyt by acceleration of SERCA activity, whilst rises in [Ca(2+)]cyt evoked by the stronger platelet activator thrombin are limited by PKC through acceleration of both SERCA and Na(+)/K(+)-ATPase activity, with the latter limiting the effect of thrombin on rises in [Na(+)]cyt and so forward mode NCX activity. The use of selective PKC inhibitors indicated that conventional and not novel PKC isoforms are responsible for the inhibition of agonist-evoked Ca(2+) signalling.


Asunto(s)
Plaquetas/metabolismo , Señalización del Calcio , Calcio/metabolismo , Proteína Quinasa C/metabolismo , Adenosina Difosfato/metabolismo , Plaquetas/efectos de los fármacos , Canales de Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Membrana Celular/metabolismo , Citosol/metabolismo , Humanos , Indoles/farmacología , Isoenzimas/metabolismo , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Isoformas de Proteínas/metabolismo , Proteína Quinasa C/genética , Trombina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA