Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
Add more filters










Publication year range
1.
J Thromb Haemost ; 9(3): 540-51, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21143372

ABSTRACT

BACKGROUND: Most Ca(2+) signaling research in platelets has relied solely on monitoring the cytosolic Ca(2+) concentration ([Ca(2+)](cyt)). Changes in [Ca(2+)](cyt) constitute the net effect of Ca(2+) fluxes into the cytosol across the plasma membrane (PM) and from intracellular stores, and Ca(2+) sequestration into the stores and Ca(2+) removal across the PM. This makes interpretation of the effects of pharmacologic or genetic interventions on Ca(2+) signaling difficult and subject to error. OBJECTIVES: To validate the use of the low-affinity Ca(2+) indicator Fluo-5N to monitor the concentration of Ca(2+) in the intracellular stores ([Ca(2+)](st)) of human platelets as a first step in developing assays for a systems-level analysis of platelet Ca(2+) signaling. METHODS: Fluo-5N-loaded and Fura-2-loaded human platelets were used to observe the effects of agonist stimulation and other manipulations on [Ca(2+)](cyt) and [Ca(2+)](st). RESULTS: Fluo-5N fluorescence changed appropriately in response to compounds that induce passive depletion of intracellular Ca(2+) stores and to physiologic agonists. Ca(2+) reuptake inhibitors and blockers of Ca(2+) release channels had the expected effects on Fura-2 and Fluo-5N fluorescence. Agonist-evoked Ca(2+) release was reversed by Ca(2+) addition to the medium, and required intact Ca(2+) reuptake mechanisms. Store refilling was observed in the presence of sarcoplasmic/endoplasmic reticulum Ca(2+) -ATPase (SERCA) inhibitors and ionomycin, suggesting the presence of a non-SERCA Ca(2+) reuptake mechanism. Evidence for a role for Ca(2+) -induced Ca(2+) release in agonist-evoked responses was obtained. CONCLUSIONS: Our data provide a validation of the use of Fluo-5N as a method for monitoring changes in [Ca(2+)](st) in human platelets.


Subject(s)
Blood Platelets/drug effects , Blood Platelets/metabolism , Calcium/blood , Calcium Signaling/drug effects , Fluorescent Dyes , Fura-2 , Humans , Hydroquinones/pharmacology , In Vitro Techniques , Inositol 1,4,5-Trisphosphate Receptors/blood , Intracellular Fluid/metabolism , Ionomycin/pharmacology , Microscopy, Confocal , NADP/analogs & derivatives , NADP/blood , Nigericin/pharmacology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Thapsigargin/pharmacology , Thrombin/pharmacology
2.
J Thromb Haemost ; 8(7): 1604-13, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20345709

ABSTRACT

SUMMARY BACKGROUND: Platelets are reported to possess a protein kinase C (PKC)-dependent non-capacitative Ca(2+)entry (NCCE) pathway. The phorbol ester, phorbol, 12-myristate, 13-acetate (PMA) has been suggested to stimulate platelet NCCE. Recently we demonstrated important roles in store-operated Ca(2+)entry in human platelets for Na(+)/Ca(2+) exchangers (NCXs) and autocrine signaling between platelets after dense granule secretion. As PMA evokes dense granule secretion, we have investigated the role of NCXs and autocrine signaling in PMA-evoked Ca(2+)entry. OBJECTIVES: To investigate the roles of NCXs and dense granule secretion in PMA-evoked Ca(2+)signaling in human platelets. METHODS: Fura-2- or sodium-binding benzofuran isophthalate (SBFI)-loaded platelets were used to monitor cytosolic Ca(2+)or Na(+) concentrations. Dense granule secretion was monitored as ATP release using luciferin-luciferase. RESULTS: The NCX inhibitors KB-R7943 or SN-6, and removal of extracellular Na(+), significantly reduced PMA-evoked Ca(2+)entry. PMA-evoked dense granule secretion was almost abolished by pretreatment with the PKC inhibitor Ro-31-8220 and significantly slowed by KB-R7943. The P(2X1) antagonists Ro-0437626 or MRS-2159, or desensitization of P(2X1) receptors by prior treatment with alpha,beta-Methylene-ATP or omitting apyrase from the medium, reduced PMA-evoked Ca(2+)entry. Ro-0437626 or chelation of extracellular Ca(2+) slowed but did not abolish PMA-evoked ATP release, indicating that PMA-evoked dense granule secretion does not require P(2X1) receptor activation but is accelerated by P(2X1)-mediated Ca(2+)entry. The presence of NCX3 in human platelets was demonstrated by Western blotting. CONCLUSION: PMA-evoked Ca(2+)entry results from an NCX3-dependent dense granule secretion and subsequent P(2X1) receptor activation by secreted ATP, rather than activation of a novel NCCE pathway.


Subject(s)
Autocrine Communication/physiology , Blood Platelets/metabolism , Calcium Signaling , Receptors, Purinergic P2X1/metabolism , Adenosine Triphosphate , Blood Platelets/drug effects , Calcium/metabolism , Calcium Signaling/drug effects , Humans , Phorbol Esters/pharmacology , Secretory Vesicles , Sodium-Calcium Exchanger/physiology
3.
J Thromb Haemost ; 7(2): 330-8, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19036069

ABSTRACT

BACKGROUND: Platelets play a role in a number of inflammatory conditions including atherosclerosis; however, the mechanisms of platelet activation under these conditions are unclear. OBJECTIVES: To investigate the presence of the vanilloid receptor, TRPV1, which is stimulated by noxious stimuli and by inflammatory mediators, in human platelets. METHODS: Platelets loaded with fura-2 or sodium-binding benzofuran isophalate acetoxymethyl ester (SBFI) were used to monitor cytosolic calcium or sodium concentrations. 5-HT secretion was determined by fluorescence assay after conjugation with o-phthaldialdehyde. ATP secretion was determined using luciferin-luciferase. RESULTS: TRPV1 was identified by Western blotting using a specific anti-hTRPV1 antibody. The TRPV1 agonist, capsaicin, evoked both Ca(2+) influx and Ca(2+) release from intracellular stores, responses that were blocked in a dose-dependent manner by the TRPV1 antagonists, 5'-Iodo-resiniferatoxin (5'-Iodo-RTX) and AMG 9810. Capsaicin also increased platelet cytosolic [Na(+)]. Capsaicin-evoked Ca(2+) release was abolished in the absence of extracellular Na(+) or by the 5-HT(2A) receptor antagonist, ketanserin. Capsaicin evoked 5-HT release from platelets, a response abolished in the absence of extracellular Na(+) or by 5'-Iodo-RTX. Thus capsaicin-evoked Ca(2+) release appeared to be mediated by Na(+)-dependent 5-HT release. TRPV1-dependent 5-HT release also contributed to ADP- and thrombin-evoked Ca(2+) entry and release. 5'-Iodo-RTX reduced ADP- and thrombin-evoked Ca(2+) signals, effects not additive with those of ketanserin, and 5'-Iodo-RTX inhibited agonist-evoked 5-HT and ATP release. CONCLUSION: These results indicate that TRPV1 is present and functionally important in human platelets. The presence of this receptor may provide a link between inflammatory mediators and platelet activation in conditions such as atherosclerosis.


Subject(s)
Platelet Activation , TRPV Cation Channels/physiology , Adenosine Triphosphate/metabolism , Blood Platelets/metabolism , Calcium/metabolism , Cytosol/metabolism , Humans , Receptor, Serotonin, 5-HT2A/metabolism , Serotonin/metabolism , Sodium/metabolism
4.
J. physiol. biochem ; 64(3): 169-178, jul.-sept. 2008. ilus, graf
Article in English | IBECS | ID: ibc-61821

ABSTRACT

Analysis of the posttranslational modification of proteins, such as phosphorylation,might yield misleading results due to the presence of other proteins with similar electrophoreticproperties that coimmunoprecipitate with the target protein. The aim ofthe present work was to develop a reliable, easy and economical technique to completelyisolate a protein from its complex. Here we present a new assay developed tofully isolate proteins from macromolecular complexes that consists of an initialSDS/PAGE (under reducing conditions), which isolates the target protein, followedby transfer of the proteins to a buffer, from which the target protein is recaptured byconventional immunoprecipitation. This technique, that we have termed “ProteinComplex Immunological Separation Assay” (ProCISA), successfully separated proteinsof different sizes, such as pp60Src and the IP3 receptor (IP3R), from their complexes.We show that ProCISA allows the investigation of the tyrosine phosphorylationstate of isolated proteins. This technique could also be used to study other posttranslationalmodifications without risk of misleading results resulting from contaminationwith other proteins of similar electrophoretic mobility which complex with theprotein of interest (AU)


No disponible


Subject(s)
Humans , Animals , Electrophoresis, Polyacrylamide Gel/methods , Immunoprecipitation/methods , Multiprotein Complexes/isolation & purification , Proteins/isolation & purification , Blotting, Western/methods , Inositol 1,4,5-Trisphosphate Receptors/chemistry , Inositol 1,4,5-Trisphosphate Receptors/isolation & purification , Oncogene Protein pp60(v-src)/chemistry , Thrombin/chemistry , Multiprotein Complexes/chemistry , Oncogene Protein pp60(v-src)/isolation & purification , Oncogene Protein pp60(v-src)/metabolism , Platelet Activation , Protein Processing, Post-Translational , Thrombin/isolation & purification , Thrombin/metabolism
6.
J Physiol Biochem ; 64(3): 169-77, 2008 Sep.
Article in English | MEDLINE | ID: mdl-19244930

ABSTRACT

Analysis of the posttranslational modification of proteins, such as phosphorylation, might yield misleading results due to the presence of other proteins with similar electrophoretic properties that coimmunoprecipitate with the target protein. The aim of the present work was to develop a reliable, easy and economical technique to completely isolate a protein from its complex. Here we present a new assay developed to fully isolate proteins from macromolecular complexes that consists of an initial SDS/PAGE (under reducing conditions), which isolates the target protein, followed by transfer of the proteins to a buffer, from which the target protein is recaptured by conventional immunoprecipitation. This technique, that we have termed "Protein Complex Immunological Separation Assay" (ProCISA), successfully separated proteins of different sizes, such as pp60Src and the IP3 receptor (IP3R), from their complexes. We show that ProCISA allows the investigation of the tyrosine phosphorylation state of isolated proteins. This technique could also be used to study other posttranslational modifications without risk of misleading results resulting from contamination with other proteins of similar electrophoretic mobility which complex with the protein of interest.


Subject(s)
Electrophoresis, Polyacrylamide Gel/methods , Immunoprecipitation/methods , Multiprotein Complexes/isolation & purification , Proteins/isolation & purification , Animals , Blotting, Western , Humans , Inositol 1,4,5-Trisphosphate Receptors/chemistry , Inositol 1,4,5-Trisphosphate Receptors/isolation & purification , Multiprotein Complexes/chemistry , Oncogene Protein pp60(v-src)/chemistry , Oncogene Protein pp60(v-src)/isolation & purification , Platelet Activation , Protein Processing, Post-Translational , Thrombin
7.
J Thromb Haemost ; 5(12): 2476-83, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17892531

ABSTRACT

BACKGROUND: Canonical transient receptor potential channels (TRPCs), which are regulated by several processes, including tyrosine phosphorylation, are candidates for the conduction of store-operated Ca(2+) entry (SOCE). OBJECTIVES: To assess hTRPC phosphotyrosine content upon platelet stimulation. METHODS: A new protein complex immunological separation assay (ProCISA) was developed to allow assessment of isolated hTRPC tyrosine phosphorylation by Western blotting. RESULTS: Classical immunoprecipitation suggested that thrombin (Thr) evoked an initial decrease in hTRPC1 phosphotyrosine content, which reached a minimum at 1 s, and then increased again, exceeding basal levels after 3 min. However, TRPC isolation from protein complexes using ProCISA revealed that hTRPC1, 4 and 5 were not tyrosine phosphorylated at rest or after Thr stimulation. Stimulation with Thr for 3 min increased the phosphotyrosine content of alpha-actinin, which shows similar electrophoretic properties to hTRPCs and coimmunoprecipitates with hTRPC1. Thr-evoked alpha-actinin tyrosine phosphorylation was increased by inhibiting the alpha-actinin phosphatase, SHP-1, which enhanced phosphorylation of the TRPC complex and SOCE. Inhibition of tyrosine phosphorylation impaired the interaction between hTRPC1 and the intracellular Ca(2+) sensor STIM1. CONCLUSIONS: hTRPC1, 4 and 5 are not tyrosine phosphorylated during SOCE in human platelets although tyrosine phosphorylation is important for SOCE. The results obtained using ProCISA caution the use of classical immunoprecipitation for the determination of the tyrosine phosphorylation state of a given protein, where the presence of other proteins with similar electrophoretic mobilities may give misleading results.


Subject(s)
Actinin/metabolism , Blood Platelets/metabolism , Calcium Signaling , Platelet Activation , TRPC Cation Channels/metabolism , Blood Platelets/drug effects , Blood Platelets/enzymology , Blotting, Western , Calcium Signaling/drug effects , Enzyme Inhibitors/pharmacology , Humans , Immunoprecipitation/methods , In Vitro Techniques , Membrane Proteins/metabolism , Multiprotein Complexes/metabolism , Neoplasm Proteins/metabolism , Phosphorylation , Phosphotyrosine/metabolism , Platelet Activation/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Protein-Tyrosine Kinases/metabolism , Quinolines/pharmacology , Stromal Interaction Molecule 1 , Thrombin/metabolism , Time Factors , Tyrosine/metabolism
8.
J Thromb Haemost ; 4(12): 2695-703, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16965542

ABSTRACT

BACKGROUND: The role of the tyrosine kinase pp60src in PAR-1-dependent Ca2+ entry was investigated in human platelets. pp60src plays a role in thapsigargin (TG)-evoked store-operated Ca2+ entry (SOCE), which is thought to be a major component of thrombin-evoked Ca2+ entry. METHODS: pp60src tyr416 phosphorylation was used to assess pp60src activation. Fura-2-loaded platelets were used to monitor intracellular Ca2+ concentration ([Ca2+]i). RESULTS: Activation of PAR-1 with the specific agonist SFLLRN increased pp60src activation within 10 s. This required phospholipase C (PLC) activity, Ca2+ release and a rise in intracellular Ca2+. PP2, an inhibitor of Src-family tyrosine kinases, inhibited SFLLRN-evoked Ca2+ entry, but also inhibited Ca2+ release and the extrusion of Ca2+ by the plasma membrane Ca2+ ATPase. Actin polymerization and conventional protein kinase C (cPKC) activity were required for TG- and SFLLRN-evoked pp60src activation. Although Gö6976, an inhibitor of cPKCs, inhibited TG-evoked SOCE, it had little effect on SFLLRN- or thrombin-evoked Ca2+ entry. CONCLUSIONS: These data indicate that stimulation of PAR-1 leads to activation of pp60src in human platelets, through PLC and cPKC activation, Ca2+ release and actin polymerization. However, as PKC and actin polymerization are not needed for SFLLRN-evoked Ca2+ entry, we suggest that pp60src is also not required. The apparent inhibition of SFLLRN-evoked Ca2+ entry by PP2 is likely to be secondary to reduced Ca2+ release. These data argue against a contribution of this SOCE pathway to PAR-1-dependent Ca2+ entry.


Subject(s)
Blood Platelets/metabolism , Calcium Signaling , Protein Kinase C/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Receptor, PAR-1/metabolism , Actins/metabolism , Blood Platelets/drug effects , Calcium/metabolism , Calcium Signaling/drug effects , Carbazoles/pharmacology , Chelating Agents/pharmacology , Cytochalasin D/pharmacology , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Humans , In Vitro Techniques , Indoles/pharmacology , Peptide Fragments/pharmacology , Phospholipid Ethers/pharmacology , Phosphorylation , Plasma Membrane Calcium-Transporting ATPases/antagonists & inhibitors , Plasma Membrane Calcium-Transporting ATPases/metabolism , Protein Kinase C/antagonists & inhibitors , Proto-Oncogene Proteins pp60(c-src)/antagonists & inhibitors , Pyrimidines/pharmacology , Receptor, PAR-1/agonists , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Thapsigargin/pharmacology , Time Factors , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism
9.
Cell Calcium ; 30(5): 323-9, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11733938

ABSTRACT

It has been reported that store-mediated Ca2+ entry (SMCE) in human platelets is likely to be mediated by a secretion-like coupling mechanism. Recently, 2-aminoethoxydiphenylborate (2-APB) has been used in the investigation of SMCE. Here, the mechanism of action of 2-APB is investigated in human platelets. In a Ca2+-free medium (EGTA added), addition of 0.1 U/ml thrombin caused an elevation in [Ca2+]i. Preincubation with 100 microM 2-APB for 170s abolished the release of internal Ca2+. In platelets whose internal Ca2+ stores had been depleted by treatment with 200 nM thapsigargin, addition of extracellular Ca2+ caused an elevation in [Ca2+]i indicative of SMCE. Preincubation with 2-APB decreased SMCE by 95.5+/-1.1%. After activation of SMCE, addition of 2-APB rapidly decreased [Ca2+]i to basal levels; in contrast, the coupling between Trp1 and IP3RII, which has been shown to play an important role in SMCE in platelets, remained intact at the same time points. The rate of decrease of [Ca2+]i and the absence of measurable latency in the effect of 2-APB were comparable to the effects of La3+ (a cation channel blocker). These data suggest that 2-APB may act as a blocker of Ca2+ permeable plasma membrane channels. These data provide further information regarding the mechanism and site of action of 2-APB and highlight the necessity of careful interpretation of work performed using this molecule.


Subject(s)
Blood Platelets/metabolism , Boron Compounds/pharmacology , Calcium Channels/drug effects , Calcium/metabolism , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Biological Transport , Blotting, Western , Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Fura-2 , Humans , Inositol 1,4,5-Trisphosphate Receptors , Lanthanum/metabolism , Precipitin Tests , Receptors, Cytoplasmic and Nuclear/metabolism , TRPC Cation Channels , Thapsigargin/pharmacology , Thrombin/pharmacology
10.
J Physiol ; 535(Pt 3): 625-35, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11559762

ABSTRACT

1. Human platelets respond to agonists of G protein (G(q))-coupled receptors by generating an irregular pattern of spiking changes in cytosolic Ca2+ ([Ca2+]i). We have investigated the ADP-induced Ca2+ responses of single, Fluo-3-loaded platelets in the presence or absence of autologous plasma or whole blood under flow conditions. 2. In plasma-free platelets, incubated in buffer medium, baseline separated [Ca2+]i peaks always consisted of a rapid rising phase (median time 0.8 s) which was abruptly followed by a slower, mono-exponential decay phase. The decay constant differed from platelet to platelet, ranging from 0.23 +/- 0.02 to 0.63 +/- 0.03 s(-1) (mean +/- S.E.M., n = 3-5), and was used to identify individual Ca2+ release events and to determine the Ca2+ fluxes of the events. 3. Confocal, high-frequency measurements of adherent, spread platelets (diameter 3-5 microm) indicated that different optical regions had simultaneous patterns of both low- and high-amplitude Ca2+ release events. 4. With or without plasma or flowing blood, the ADP-induced Ca2+ signals in platelets had the characteristics of irregular Ca2+ puffs as well as more regular Ca2+ oscillations. Individual [Ca2+]i peaks varied in amplitude and peak-to-peak interval, as observed for separated Ca2+ puffs within larger cells. On the other hand, the peaks appeared to group into periods of ragged, shorter-interval Ca2+ release events with little integration, which were alternated with longer-interval events. 5. We conclude that the spiking Ca2+ signal generated in these small cells has the characteristics of a 'poor' oscillator with an irregular frequency being reactivated from period to period. This platelet signal appears to be similar in an environment of non-physiological buffer medium and in flowing, whole blood.


Subject(s)
Adenosine Diphosphate/pharmacology , Blood Platelets/metabolism , Calcium Signaling/drug effects , Calcium/blood , Blood Platelets/drug effects , Buffers , Calibration , Culture Media , GTP-Binding Proteins/metabolism , HeLa Cells , Humans , In Vitro Techniques , Microscopy, Confocal , Microscopy, Fluorescence , Microscopy, Video , Stimulation, Chemical
11.
Am J Physiol Cell Physiol ; 280(6): C1636-44, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11350760

ABSTRACT

Tumor necrosis factor-alpha (TNF-alpha) is an important component of the early signaling pathways leading to liver regeneration and proliferation, but it is also responsible for several hepatotoxic effects. We have investigated the effect of TNF-alpha on thapsigargin (TG)-induced store-mediated Ca2+ entry (SMCE) in the human hepatocellular carcinoma cell line HepG2. In these cells, short-term (10 min) exposure to TNF-alpha slightly increased SMCE. In contrast, long-term (12 h) exposure to TNF-alpha significantly reduced SMCE. This effect was reversed by coincubation with atrial natriuretic peptide (ANP), which itself had no effect on SMCE. Cytochalasin D and latrunculin A, inhibitors of actin polymerization, abolished SMCE. Long-term exposure of HepG2 cells to TNF-alpha abolished TG-induced actin polymerization and membrane association of Ras proteins. When TNF-alpha was added in combination with ANP, these effects were reduced. These findings suggest that in HepG2 cells, TNF-alpha inhibits SMCE by affecting reorganization of the actin cytoskeleton, probably by interfering with the activation of Ras proteins, and that ANP protects against these inhibitory effects of TNF-alpha.


Subject(s)
Calcium/metabolism , Carcinoma, Hepatocellular , Liver Neoplasms , Tumor Necrosis Factor-alpha/pharmacology , Actins/metabolism , Atrial Natriuretic Factor/analysis , Atrial Natriuretic Factor/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Carcinogens/pharmacology , Cell Compartmentation/drug effects , Cell Compartmentation/physiology , Cytochalasin D/pharmacology , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Humans , Nucleic Acid Synthesis Inhibitors/pharmacology , Polymers/metabolism , Thapsigargin/pharmacology , Thiazoles/pharmacology , Thiazolidines , Tumor Cells, Cultured/chemistry , Tumor Cells, Cultured/metabolism , ras Proteins/analysis
12.
Biochem J ; 356(Pt 1): 191-8, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11336651

ABSTRACT

Physical coupling between inositol 1,4,5-trisphosphate (IP(3)) receptors and transient receptor potential (Trp) channels has been demonstrated in both transfected and normal cells as a candidate mechanism for the activation of store-mediated Ca(2+) entry (SMCE). We have investigated the properties of the coupling between the type II IP(3) receptor and naturally expressed human Trp1 (hTrp1) in human platelets. Treatment with xestospongin C, an inhibitor of IP(3) receptor function, abolished SMCE and coupling between the IP(3) receptor and hTrp1. The coupling was activated by depletion of the intracellular Ca(2+) stores, and was reversed by refilling of the stores. We have also examined the role of actin filaments in the activation and maintenance of the coupling. Stabilization of the cortical actin network with jasplakinolide prevented the coupling, indicating that, as with secretion, the actin filaments at the cell periphery act as a negative clamp which prevents constitutive coupling. In addition, the actin cytoskeleton plays a positive role, since disruption of the actin network inhibited the coupling when the Ca(2+) stores were depleted. These results provide strong evidence for the activation of SMCE by a secretion-like coupling mechanism involving a reversible association between IP(3) receptors and hTrp1 in normal human cells.


Subject(s)
Blood Platelets/metabolism , Calcium Channels/metabolism , Calcium/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Actins/metabolism , Biological Transport , Cytochalasin D/pharmacology , Cytoskeleton/metabolism , Enzyme Inhibitors/pharmacology , Humans , Inositol 1,4,5-Trisphosphate Receptors , Macrocyclic Compounds , Oxazoles/pharmacology , Protein Binding , Receptor Cross-Talk , TRPC Cation Channels
13.
Blood ; 97(9): 2648-56, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11313254

ABSTRACT

Effects of the occupation of integrin alpha(IIb)beta(3) by fibrinogen on Ca(++) signaling in fura-2-loaded human platelets were investigated. Adding fibrinogen to washed platelet suspensions inhibited increases in cytosolic [Ca(++)] concentrations ([Ca(++)](i)) evoked by adenosine diphosphate (ADP) and thrombin in a concentration-dependent manner in the presence of external Ca(++) but not in the absence of external Ca(++) or in the presence of the nonselective cation channel blocker SKF96365, indicating selective inhibition of Ca(++) entry. Fibrinogen also inhibited store-mediated Ca(++) entry (SMCE) activated after Ca(++) store depletion using thapsigargin. The inhibitory effect of fibrinogen was reversed if fibrinogen binding to alpha(IIb)beta(3) was blocked using RDGS or abciximab and was absent in platelets from patients homozygous for Glanzmann thrombasthenia. Fibrinogen was without effect on SMCE once activated. Activation of SMCE in platelets occurs through conformational coupling between the intracellular stores and the plasma membrane and requires remodeling of the actin cytoskeleton. Fibrinogen inhibited actin polymerization evoked by ADP or thapsigargin in control cells and in cells loaded with the Ca(++) chelator dimethyl BAPTA. It also inhibited the translocation of the tyrosine kinase p60(src) to the cytoskeleton. These results indicate that the binding of fibrinogen to integrin alpha(IIb)beta(3) inhibits the activation of SMCE in platelets by a mechanism that may involve modulation of the reorganization of the actin cytoskeleton and the cytoskeletal association of p60(src). This action may be important in intrinsic negative feedback to prevent the further activation of platelets subjected.


Subject(s)
Blood Platelets/metabolism , Calcium/metabolism , Fibrinogen/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Fibrinogen/chemistry , Humans , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Protein Binding , Signal Transduction , Thrombasthenia/blood
14.
J Biol Chem ; 276(19): 15666-75, 2001 May 11.
Article in English | MEDLINE | ID: mdl-11278478

ABSTRACT

Agonists elevate the cytosolic calcium concentration in human platelets via a receptor-operated mechanism, involving both Ca(2+) release from intracellular stores and subsequent Ca(2+) entry, which can be inhibited by platelet inhibitors, such as prostaglandin E(1) and nitroprusside which elevate cAMP and cGMP, respectively. In the present study we investigated the mechanisms by which cAMP and cGMP modulate store-mediated Ca(2+) entry. Both prostaglandin E(1) and sodium nitroprusside inhibited thapsigargin-evoked store-mediated Ca(2+) entry and actin polymerization. However, addition of these agents after induction of store-mediated Ca(2+) entry did not affect either Ca(2+) entry or actin polymerization. Furthermore, prostaglandin E(1) and sodium nitroprusside dramatically inhibited the tyrosine phosphorylation induced by depletion of the internal Ca(2+) stores or agonist stimulation without affecting the activation of Ras or the Ras-activated phosphatidylinositol 3-kinase or extracellular signal-related kinase (ERK) pathways. Inhibition of cyclic nucleotide-dependent protein kinases prevented inhibition of agonist-evoked Ca(2+) release but it did not have any effect on the inhibition of Ca(2+) entry or actin polymerization. Phenylarsine oxide and vanadate, inhibitors of protein-tyrosine phosphatases prevented the inhibitory effects of the cGMP and cAMP elevating agents on Ca(2+) entry and actin polymerization. These results suggest that Ca(2+) entry in human platelets is directly down-regulated by cGMP and cAMP by a mechanism involving the inhibition of cytoskeletal reorganization via the activation of protein tyrosine phosphatases.


Subject(s)
Actins/blood , Blood Platelets/physiology , Calcium/blood , Carbazoles , Cyclic AMP/blood , Cyclic GMP/blood , Mitogen-Activated Protein Kinases/blood , Platelet Aggregation Inhibitors/pharmacology , Protein Tyrosine Phosphatases/blood , Actins/chemistry , Adenosine Diphosphate/pharmacology , Alprostadil/pharmacology , Arsenicals/pharmacology , Blood Platelets/drug effects , Egtazic Acid/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , In Vitro Techniques , Indoles/pharmacology , Mitogen-Activated Protein Kinase 1/blood , Mitogen-Activated Protein Kinase 3 , Nitroprusside/pharmacology , Phosphorylation , Platelet Activation/drug effects , Platelet Activation/physiology , Protein Tyrosine Phosphatases/antagonists & inhibitors , Pyrroles/pharmacology , Thapsigargin/pharmacology , Thrombin/pharmacology , Vanadates/pharmacology
15.
J Biol Chem ; 276(19): 15659-65, 2001 May 11.
Article in English | MEDLINE | ID: mdl-11278479

ABSTRACT

Extracellular signal-regulated kinases (ERKs), are common participants in a broad variety of signal transduction pathways. Several studies have demonstrated the presence of ERKs in human platelets and their activation by the physiological agonist thrombin. Here we report the involvement of the ERK cascade in store-mediated Ca(2+) entry in human platelets. Treatment of dimethyl-bis-(o-aminophenoxy)-ethane-N,N,N',N'-tetraacetic acid-loaded platelets with thapsigargin to deplete the intracellular Ca(2+) stores resulted in a time- and concentration-dependent activation of ERK1 and ERK2. Incubation with either U0126 or PD 184352, specific inhibitors of mitogen-activated protein kinase kinase (MEK), prevented thapsigargin-induced ERK activation. Furthermore, U0126 and PD 184352 reduced Ca(2+) entry stimulated by thapsigargin or thrombin, in a concentration-dependent manner. The role of ERK in store-mediated Ca(2+) entry was found to be independent of phosphatidylinositol 3- and 4-kinases, the tyrosine kinase pathway, and actin polymerization but sensitive to treatment with inhibitors of Ras, suggesting that the ERK pathway might be a downstream effector of Ras in mediating store-mediated Ca(2+) entry in human platelets. In addition, we have found that store depletion stimulated ERK activation does not require PKC activity. This study demonstrates for the first time a novel mechanism for regulation of store-mediated Ca(2+) entry in human platelets involving the ERK cascade.


Subject(s)
Blood Platelets/physiology , Calcium/blood , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/blood , Platelet Activation , Actins/blood , Benzamides/pharmacology , Blood Platelets/drug effects , Butadienes/pharmacology , Cytochalasin D/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Farnesol/analogs & derivatives , Farnesol/pharmacology , Humans , In Vitro Techniques , Kinetics , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/blood , Mitogen-Activated Protein Kinase 3 , Nitriles/pharmacology , Thapsigargin/pharmacology , Thrombin/pharmacology
16.
J Physiol ; 529 Pt 1: 159-69, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11080259

ABSTRACT

1. In many non-excitable cells Ca2+ influx is mainly controlled by the filling state of the intracellular Ca2+ stores. It has been suggested that this store-mediated or capacitative Ca2+ entry is brought about by a physical and reversible coupling of the endoplasmic reticulum with the plasma membrane. Here we provide evidence for an additional, non-capacitative Ca2+ entry mechanism in human platelets. 2. Changes in cytosolic Ca2+ and Sr2+ were measured in human platelets loaded with the fluorescent indicator fura-2. 3. Depletion of the internal Ca2+ stores with thapsigargin plus a low concentration of ionomycin stimulated store-mediated cation entry, as demonstrated upon Ca2+ or Sr2+ addition. Subsequent treatment with thrombin stimulated further divalent cation entry in a concentration-dependent manner. 4. Direct activation of protein kinase C (PKC) by phorbol-12-myristate-13-acetate or 1-oleoyl-2-acetyl-sn-glycerol also stimulated divalent cation entry, without evoking the release of Ca2+ from intracellular stores. Cation entry evoked by thrombin or activators of PKC was abolished by the PKC inhibitor Ro-31-8220. 5. Unlike store-mediated Ca2+ entry, jasplakinolide, which reorganises actin filaments into a tight cortical layer adjacent to the plasma membrane, did not inhibit divalent cation influx evoked by thrombin when applied after Ca2+ store depletion, or by activators of PKC. Thrombin also activated Ca2+ entry in platelets in which the release from intracellular stores and store-mediated Ca2+ entry were blocked by xestospongin C. 6. These results indicate that the non-capacitative divalent cation entry pathway is regulated independently of store-mediated entry and does not require coupling of the endoplasmic reticulum and the plasma membrane. These results support the existence of a mechanism for receptor-evoked Ca2+ entry in human platelets that is independent of Ca2+ store depletion. This Ca2+ entry mechanism may be activated by occupation of G-protein-coupled receptors, which activate PKC, or by direct activation of PKC, thus generating non-capacitative Ca2+ entry alongside that evoked following the release of Ca2+ from the intracellular stores.


Subject(s)
Blood Platelets/metabolism , Calcium Channels/blood , Depsipeptides , Protein Kinase C/metabolism , Blood Platelets/drug effects , Calcium/blood , Calcium Channels/drug effects , Cations, Divalent/pharmacology , Enzyme Inhibitors/pharmacology , Humans , In Vitro Techniques , Indoles/pharmacology , Macrocyclic Compounds , Microscopy, Confocal , Oxazoles/pharmacology , Peptides, Cyclic/pharmacology , Protein Kinase C/antagonists & inhibitors , Receptors, Purinergic P2/drug effects , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2X , Strontium/blood , Thrombin/pharmacology
17.
Biochem J ; 351 Pt 2: 429-37, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11023829

ABSTRACT

We have recently reported that store-mediated Ca(2+) entry in platelets is likely to be mediated by a reversible trafficking and coupling of the endoplasmic reticulum with the plasma membrane, a model termed 'secretion-like coupling'. In this model the actin cytoskeleton plays a key regulatory role. Since tyrosine kinases have been shown to be important for Ca(2+) entry in platelets and other cells, we have now investigated the possible involvement of tyrosine kinases in the secretion-like-coupling model. Treatment of platelets with thrombin or thapsigargin induced actin polymerization by a calcium-independent pathway. Methyl 2,5-dihydroxycinnamate, a tyrosine kinase inhibitor, prevented thrombin- or thapsigargin-induced actin polymerization. The effects of tyrosine kinases in store-mediated Ca(2+) entry were found to be entirely dependent on the actin cytoskeleton. PP1, an inhibitor of the Src family of proteins, partially inhibited store-mediated Ca(2+) entry. In addition, depletion of intracellular Ca(2+) stores stimulated cytoskeletal association of the cytoplasmic tyrosine kinase pp60(src), a process that was sensitive to treatment with cytochalasin D and PP1, but not to inhibition of Ras proteins using prenylcysteine analogues. Finally, combined inhibition of both Ras proteins and tyrosine kinases resulted in complete inhibition of Ca(2+) entry, suggesting that these two families of proteins have independent effects in the activation of store-mediated Ca(2+) entry in human platelets.


Subject(s)
Actins/metabolism , Blood Platelets/metabolism , Calcium/metabolism , Cytoskeleton/metabolism , Protein-Tyrosine Kinases/physiology , Cinnamates/pharmacology , Cytochalasin D/pharmacology , Dose-Response Relationship, Drug , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Ions , Nucleic Acid Synthesis Inhibitors/pharmacology , Phosphorylation , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Proteins/pharmacology , Proto-Oncogene Proteins pp60(c-src)/metabolism , Thapsigargin/pharmacology , Thrombin/pharmacology , Time Factors , Tyrosine/metabolism , ras Proteins/metabolism
18.
Cell Calcium ; 28(2): 119-26, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10970768

ABSTRACT

The roles of P(2X1)and P(2T AC)receptors in ADP-evoked Ca(2+)signalling were investigated in fura-2-loaded human platelets. Desensitization of the P(2X1)receptor with the selective agonist, alphabeta-methylene ATP, reduced the integral of the ADP-evoked rise in [Ca(2+)](i)to about 90% of control; a reduction equivalent to the integral of the P(2X1)-evoked response alone. After elevating cAMP or cGMP levels using prostaglandin E(1)or sodium nitroprusside, prior P(2X1)desensitization reduced the integral of the ADP-evoked response to about 70% of control. This reduction was greater than the integral of the P(2X1)-evoked response alone under the same conditions, suggesting rapidly activated Ca(2+)entry via the P(2X1)receptor potentiates Ca(2+)responses evoked via the phospholipase C-coupled P(2Y1)receptor. The P(2T AC)receptor antagonist, AR-C69931MX, at a concentration completely inhibiting aggregation, did not significantly affect the initial peaks but caused a significant reduction in the integrals of the ADP-evoked rises in [Ca(2+)](i)to about 71% or 77% of controls in the presence or absence of external Ca(2+)respectively. This suggests that the main effect of lowering cAMP levels after inhibition of adenylyl cyclase via P(2T AC)receptors may be reduced Ca(2+)removal from the cytosol. These results indicate that both the P(2X1)and P(2T AC)receptors play a significant role in ADP-evoked Ca(2+)signalling in human platelets.


Subject(s)
Blood Platelets/metabolism , Calcium Signaling , Membrane Proteins , Receptors, Purinergic P2/metabolism , Adenosine Diphosphate/pharmacology , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Blood Platelets/drug effects , Calcium Signaling/drug effects , Humans , Platelet Aggregation Inhibitors/pharmacology , Purinergic P2 Receptor Agonists , Purinergic P2 Receptor Antagonists , Receptors, Purinergic P2X , Receptors, Purinergic P2Y12
19.
Biochem J ; 350 Pt 3: 631-5, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10970773

ABSTRACT

In the present study we have investigated the role of inositol 1,4, 5-trisphosphate (IP(3)), functional IP(3) receptors (IP(3)Rs) and the human homologue of the Drosophila transient receptor potential (Trp) channel, human Trp1 (hTrp1), in store-mediated Ca(2+) entry (SMCE) in human platelets. Inhibition of IP(3) recycling using Li(+), or the inhibition of IP(3)Rs using xestospongin C, both resulted in the inhibition of SMCE activation following Ca(2+) store depletion using thapsigargin. Co-immunoprecipitation experiments indicated that endogenously expressed hTrp1 couples with IP(3)R type II, but not types I or III, in platelets with depleted intracellular Ca(2+) stores, but not in control, undepleted cells. These results provide strong evidence for the activation of SMCE by conformational coupling involving de novo association between IP(3)Rs and a plasma membrane channel in normal human cells.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Fungal Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Blood Platelets/metabolism , Cells, Cultured , Humans , Inositol 1,4,5-Trisphosphate Receptors , Protein Binding , Protein Isoforms/metabolism
20.
Platelets ; 11(4): 215-21, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10938900

ABSTRACT

In human platelets and other non-excitable cell types depletion of the intracellular calcium stores promotes calcium entry across the plasma membrane. Although the mechanism of this store-mediated calcium entry remains uncertain, it has been suggested that a tyrosine phosphorylation step could be involved. In support of this hypothesis various tyrosine kinase inhibitors have been shown to reduce store-mediated calcium entry in platelets, although this inhibition is never complete. Here we investigate the properties of store-mediated calcium entry in human platelets during the time course of its activation. Our data suggest that at least two pathways may contribute to store-mediated calcium entry in these cells. An early component, activated soon after the initiation of Ca2+ store depletion, is insensitive to trivalent cations, SKF 96365 and tyrosine kinase inhibitors. This is followed by a second component which is inhibited by La3+, SKF 96365 and by tyrosine kinase inhibitors. These results suggest a role for tyrosine kinases in generating only the later stages of store-mediated calcium entry in platelets and may explain the incomplete inhibition of this pathway by inhibitors of tyrosine kinases.


Subject(s)
Blood Platelets/metabolism , Calcium/metabolism , Androstadienes/pharmacology , Biological Transport/drug effects , Blood Platelets/chemistry , Calcium Channel Blockers/pharmacology , Calcium Signaling/drug effects , Chelating Agents/pharmacology , Econazole/pharmacology , Egtazic Acid/pharmacology , Enzyme Inhibitors , Humans , Imidazoles/pharmacology , Ionomycin/pharmacology , Ionophores/pharmacology , Lanthanum/pharmacology , Manganese/metabolism , Phosphorylation , Protein-Tyrosine Kinases/antagonists & inhibitors , Thapsigargin/pharmacology , Tyrosine/metabolism , Wortmannin
SELECTION OF CITATIONS
SEARCH DETAIL
...