Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Cell Signal ; 26(3): 502-11, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24308967

ABSTRACT

Ca(2+) elevation is essential to platelet activation. STIM1 senses Ca(2+) in the endoplasmic reticulum and activates Orai channels allowing store-operated Ca(2+) entry (SOCE). STIM1 has also been reported to be present in the plasma membrane (PM) with its N-terminal region exposed to the outside medium but its role is not fully understood. We have examined the effects of the antibody GOK/STIM1, which recognises the N-terminal region of STIM1, on SOCE, agonist-stimulated Ca(2+) entry, surface exposure, in vitro thrombus formation and aggregation in human platelets. We also determined novel binding partners of STIM1 using proteomics. The dialysed GOK/STIM1 antibody failed to reduced thapsigargin- and agonist-mediated Ca(2+) entry in Fura2-labelled cells. Using flow cytometry we detect a portion of STIM1 to be surface-exposed. The dialysed GOK/STIM1 antibody reduced thrombus formation by whole blood on collagen-coated capillaries under flow and platelet aggregation induced by collagen. In immunoprecipitation experiments followed by proteomic analysis, STIM1 was found to extract a number of proteins including myosin, DOCK10, thrombospondin-1 and actin. These studies suggest that PM STIM1 may facilitate platelet activation by collagen through novel interactions at the plasma membrane while the essential Ca(2+)-sensing role of STIM1 is served by the protein in the ER.


Subject(s)
Collagen/metabolism , Endoplasmic Reticulum/metabolism , Membrane Proteins/immunology , Neoplasm Proteins/immunology , Platelet Activation/immunology , Platelet Aggregation/immunology , Acetamides/pharmacology , Actins/metabolism , Anilides/pharmacology , Antibodies/immunology , Antibodies/pharmacology , Blood Platelets , Calcium , Calcium Signaling/drug effects , Calcium Signaling/immunology , Enzyme Inhibitors/pharmacology , Guanine Nucleotide Exchange Factors/metabolism , Humans , Ion Channels/drug effects , Isoquinolines/pharmacology , Myosins/metabolism , Platelet Activation/drug effects , Protein Binding/immunology , Stromal Interaction Molecule 1 , Thapsigargin/pharmacology , Thiadiazoles/pharmacology , Thrombosis/immunology , Thrombospondin 1/metabolism
2.
Handb Exp Pharmacol ; (179): 425-43, 2007.
Article in English | MEDLINE | ID: mdl-17217071

ABSTRACT

Ca2+ entry forms an essential component of platelet activation; however, the mechanisms associated with this process are not understood. Ca2+ entry upon receptor activation occurs as a consequence of intracellular store depletion (referred to as store-operated Ca2+ entry or SOCE), a direct action of second messengers on cation entry channels or the direct occupancy of a ligand-gated P2(Xi) receptor. The molecular identity of the SOCE channel has yet to be established. Transient receptor potential (TRP) proteins are candidate cation entry channels and are classified into a number of closely related subfamilies including TRPC (canonical), TRPV (vanilloid), TRPM (melastatin), TRPP (polycystin) and TRPML (mucolipins). From the TRPC family, platelets have been shown to express TRPC6 and TRPC1, and are likely to express other TRPC and other TRP members. TRPC6 is suggested to be involved with receptor-activated, diacyl-glycerol-mediated cation entry. TRPC1 has been suggested to be involved with SOCE, though many of the suggested mechanisms remain controversial. As no single TRP channel has the properties described for SOCE in platelets, it is likely that it is composed of a heteromeric association of TRP and related subunits, some of which may be present in intracellular compartments in the resting cell.


Subject(s)
Blood Platelets/physiology , Transient Receptor Potential Channels/blood , Transient Receptor Potential Channels/physiology , Animals , Calcium Channels/physiology , Calcium Signaling/physiology , Gene Expression Regulation , Humans , Megakaryocytes/metabolism , Megakaryocytes/physiology , Signal Transduction/physiology , Transient Receptor Potential Channels/genetics
3.
Br J Dermatol ; 151(2): 440-5, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15327552

ABSTRACT

BACKGROUND: Darier's disease (DD) is caused by mutations in ATP2A2, which encodes the sarco/endoplasmic reticulum calcium ATPase type 2 (SERCA2), a member of a family of calcium pumps important in intracellular calcium signalling. SERCA2 has two isoforms. SERCA2a occurs mainly in cardiac and skeletal muscle, whereas SERCA2b occurs ubiquitously and is coexpressed with the related SERCA type 3 (SERCA3) in many tissues. It is not known why mutations in the widely expressed SERCA2 manifest as a focal skin disease. OBJECTIVES: To provide insight into the pathogenesis of DD by examining SERCA isoform expression in normal skin and DD skin. METHODS: Using immunohistochemistry we studied SERCA2a, SERCA2b and SERCA3 expression in nonlesional and lesional skin from seven patients with DD and normal skin from seven control subjects. We quantified SERCA2a and SERCA2b staining intensity by grey scale analysis of fluorescence intensity. RESULTS: In normal and DD epidermis both SERCA2a and SERCA2b staining was seen. SERCA2a staining in epidermis was less intense relative to pilar muscle whereas SERCA2b staining in epidermis was of marginally greater intensity than in pilar muscle. SERCA3 was not expressed in normal or DD epidermis, but was found in eccrine glands and blood vessels. No reduction was detected in SERCA2a or SERCA2b staining intensity in DD nonlesional epidermis compared with control epidermis. In within-patient comparisons, SERCA2a and SERCA2b staining in lesional epidermis was less intense than in nonlesional epidermis. CONCLUSIONS: Both SERCA2a and SERCA2b are present in epidermis, although the latter may predominate. The absence of coexpressed SERCA3 in epidermis may explain the localization of DD. Comparable SERCA2 staining intensity in nonlesional DD and control epidermis, even in patients predicted to be haploinsufficient, suggests partial compensation by upregulation of the normal allele. Unknown additional factors may trigger focal lesions by overcoming this compensation. Reduced staining intensity in lesional tissue may be secondary, or may reflect local downregulation of SERCA2 expression predisposing to development of focal lesions.


Subject(s)
Calcium-Transporting ATPases/analysis , Darier Disease/enzymology , Adult , Aged , Eccrine Glands/enzymology , Epidermis/enzymology , Humans , Immunohistochemistry/methods , Middle Aged , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Skin/blood supply
4.
Blood ; 95(11): 3412-22, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10828023

ABSTRACT

Platelet activation is associated with an increase of cytosolic Ca(++) levels. The (1,4,5)IP(3) receptors [(1,4,5)IP(3)R] are known to mediate Ca(++) release from intracellular stores of many cell types. Currently there are at least 3 distinct subtypes of (1,4, 5)IP(3)R-type I, type II, and type III-with suggestions of distinct roles in Ca(++) elevation. Specific receptors for (1,3,4,5)IP(4) belonging to the GAP1 family have also been described though their involvement with Ca(++) regulation is controversial. In this study we report that platelets contain all 3 subtypes of (1,4,5)IP(3)R but in different amounts. Type I and type II receptors are predominant. In studies using highly purified platelet plasma (PM) and intracellular membranes (IM) we report a distinct localization of these receptors. The PM fractions were found to contain the type III (1,4,5)IP(3)R and GAP1(IP4BP) in contrast to IM, which contained type I (1,4,5)IP(3)R. The type II receptor exhibited a dual distribution. In studies examining the labeling of surface proteins with biotin in intact platelets only the type III (1,4,5)IP(3)R was significantly labeled. Immunogold studies of ultracryosections of human platelets showed significantly more labeling of the PM with the type III receptor antibodies than with type I receptor antibodies. Ca(++) flux studies were carried out with the PM to demonstrate in vitro function of inositol phosphate receptors. Ca(++) release activities were present with both (1,4,5)IP(3) and (1, 3,4,5)IP(4) (EC(50) = 1.3 and 0.8 micromol/L, respectively). Discrimination of the Ca(++)-releasing activities was demonstrated with cyclic adenosine monophosphate (cAMP)-dependent protein kinase (cAMP-PK) specifically inhibiting (1,4,5)IP(3) but not (1,3,4, 5)IP(4)-induced Ca(++) flux. In experiments with both PM and intact platelets, the (1,4,5)IP(3)Rs but not GAP1(IP4BP) were found to be substrates of cAMP-PK and cGMP-PK. Thus the Ca(++) flux property of (1,3,4,5)IP(4) is insensitive to cAMP-PK. These studies suggest distinct roles for the (1,4,5)IP(3)R subtypes in Ca(++) movements, with the type III receptor and GAP1(IP4BP) associated with cation entry in human platelets and the type I receptor involved with Ca(++) release from intracellular stores.


Subject(s)
Blood Platelets/metabolism , Calcium Channels/blood , Calcium/blood , Receptors, Cytoplasmic and Nuclear/blood , Blood Platelets/chemistry , Blood Platelets/ultrastructure , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cyclic AMP-Dependent Protein Kinase Type II , Cyclic AMP-Dependent Protein Kinases/blood , Humans , Inositol 1,4,5-Trisphosphate/pharmacology , Inositol 1,4,5-Trisphosphate Receptors , Inositol Phosphates/blood , Intracellular Membranes/chemistry , Intracellular Membranes/metabolism , Intracellular Membranes/ultrastructure , Kinetics , Microscopy, Immunoelectron , Models, Biological , Phosphorylation , Protein Isoforms/blood
5.
J Biol Chem ; 275(21): 16242-50, 2000 May 26.
Article in English | MEDLINE | ID: mdl-10748064

ABSTRACT

This study presents evidence that phosphoinositide (PI) 3-kinase is involved in T cell Ca(2+) signaling via a phosphatidylinositol 3,4, 5-trisphosphate PI(3,4,5)P(3)-sensitive Ca(2+) entry pathway. First, exogenous PI(3,4,5)P(3) at concentrations close to its physiological levels induces Ca(2+) influx in T cells, whereas PI(3,4)P(2), PI(4, 5)P(2), and PI(3)P have no effect on [Ca(2+)](i). This Ca(2+) entry mechanism is cell type-specific as B cells and a number of cell lines examined do not respond to PI(3,4,5)P(3) stimulation. Second, inhibition of PI 3-kinase by wortmannin and by overexpression of the dominant negative inhibitor Deltap85 suppresses anti-CD3-induced Ca(2+) response, which could be reversed by subsequent exposure to PI(3,4,5)P(3). Third, PI(3,4,5)P(3) is capable of stimulating Ca(2+) efflux from Ca(2+)-loaded plasma membrane vesicles prepared from Jurkat T cells, suggesting that PI(3,4,5)P(3) interacts with a Ca(2+) entry system directly or via a membrane-bound protein. Fourth, although D-myo-inositol 1,3,4,5-tetrakisphosphate (Ins(1,3,4, 5)P(4)) mimics PI(3,4,5)P(3) in many aspects of biochemical functions such as membrane binding and Ca(2+) transport, we raise evidence that Ins(1,3,4,5)P(4) does not play a role in anti-CD3- or PI(3,4,5)P(3)-mediated Ca(2+) entry. This PI(3,4,5)P(3)-stimulated Ca(2+) influx connotes physiological significance, considering the pivotal role of PI 3-kinase in the regulation of T cell function. Given that PI 3-kinase and phospholipase C-gamma form multifunctional complexes downstream of many receptor signaling pathways, we hypothesize that PI(3,4,5)P(3)-induced Ca(2+) entry acts concertedly with Ins(1,4,5)P(3)-induced Ca(2+) release in initiating T cell Ca(2+) signaling. By using a biotinylated analog of PI(3,4,5)P(3) as the affinity probe, we have detected several putative PI(3,4,5)P(3)-binding proteins in T cell plasma membranes.


Subject(s)
Calcium Signaling , Phosphatidylinositol 3-Kinases/metabolism , T-Lymphocytes/enzymology , Androstadienes/pharmacology , CD3 Complex/immunology , CD3 Complex/metabolism , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Cell Membrane/metabolism , Enzyme Inhibitors/pharmacology , Fura-2 , Humans , Imidazoles/pharmacology , Indoles , Inositol 1,4,5-Trisphosphate Receptors , Inositol Phosphates/pharmacology , Jurkat Cells , Liposomes/metabolism , Phosphatidylinositol 4,5-Diphosphate/pharmacology , Phosphatidylinositols/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Wortmannin
6.
Thromb Haemost ; 79(1): 177-85, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9459345

ABSTRACT

We report the characterization of a monoclonal antibody (MAb) PM6/13 which recognises glycoprotein IIIa (GPIIIa) on platelet membranes and in functional studies inhibits platelet aggregation induced by all agonists examined. In platelet-rich plasma, inhibition of aggregation induced by ADP or low concentrations of collagen was accompanied by inhibition of 5-hydroxytryptamine secretion. EC50 values were 10 and 9 microg/ml antibody against ADP and collagen induced responses respectively. In washed platelets treated with the cyclooxygenase inhibitor, indomethacin, PM6/13 inhibited platelet aggregation induced by thrombin (0.2 U/ml), collagen (10 microg/ml) and U46619 (3 microM) with EC50 = 4, 8 and 4 microg/ml respectively, without affecting [14C]5-hydroxytryptamine secretion or [3H]arachidonate release in appropriately labelled cells. Studies in Fura 2-labelled platelets revealed that elevation of intracellular calcium by ADP, thrombin or U46619 was unaffected by PM6/13 suggesting that the epitope recognised by the antibody did not influence Ca2+ regulation. In agreement with the results from the platelet aggregation studies, PM6/13 was found to potently inhibit binding of 125I-fibrinogen to ADP activated platelets. Binding of this ligand was also inhibited by two other MAbs tested, namely SZ-21 (also to GPIIIa) and PM6/248 (to the GPIIb-IIIa complex). However when tested against binding of 125I-fibronectin to thrombin stimulated platelets, PM6/13 was ineffective in contrast with SZ-21 and PM6/248, that were both potent inhibitors. This suggested that the epitopes recognised by PM6/13 and SZ-21 on GPIIIa were distinct. Studies employing proteolytic dissection of 125I-labelled GPIIIa by trypsin followed by immunoprecipitation with PM6/13 and analysis by SDS-PAGE, revealed the presence of four fragments at 70, 55, 30 and 28 kDa. PM6/13 did not recognize any protein bands on Western blots performed under reducing conditions. However Western blotting analysis with PM6/13 under non-reducing conditions revealed strong detection of the parent GPIIIa molecule, of trypsin treated samples revealed recognition of an 80 kDa fragment at 1 min, faint recognition of a 60 kDa fragment at 60 min and no recognition of any product at 18 h treatment. Under similar conditions, SZ-21 recognized fragments at 80, 75 and 55 kDa with the 55 kDa species persisting even after 18 h trypsin treatment. These studies confirm the epitopes recognised by PM6/13 and SZ-21 to be distinct and that PM6/13 represents a useful tool to differentiate the characteristics of fibrinogen and fibronectin binding to the GPIIb-IIIa complex on activated platelets.


Subject(s)
Fibrinogen/metabolism , Fibronectins/blood , Platelet Activation , Platelet Aggregation/immunology , Platelet Glycoprotein GPIIb-IIIa Complex/immunology , Antibodies, Monoclonal , Arachidonic Acid/blood , Humans , Male , Protein Binding , Reference Values , Trypsin
7.
FEBS Lett ; 403(1): 83-6, 1997 Feb 10.
Article in English | MEDLINE | ID: mdl-9038365

ABSTRACT

Store-regulated Ca2+ entry represents a major mechanism for Ca2+ influx in non-excitable cells although many details remain to be evaluated including the identification of cation entry channels. Recently human homologues of the Drosophila proteins TRP and TRPL, have been described (TRPC1, TRPC1A, HTRP1) and suggested as candidate cation channels. In this study we sought to examine if the producers of blood platelets, megakaryocytic cells (using the cell lines MEG01, DAMI, HEL), expressed these genes. RNA was prepared from the cell lines and platelets and converted to cDNA. The cDNA was then subjected to 30-35 cycles of PCR using gene specific primers for TRPC1-3. PCR products of the expected sizes were observed for all three TRPC genes in the three cell lines. Direct sequencing confirmed their identity. Additionally for TRPC1, a larger species, and for TRPC2, a smaller species was detected in all three cell lines with sequencing revealing the fragments to contain TRPC sequence, suggesting that they were either products of alternative splicing events or from closely related genes. These results suggest that TRPC genes are expressed in megakaryocytic cell lines and that the TRPC proteins may play a role in mediating cation influx in both megakaryocytes and platelets.


Subject(s)
Calcium Channels , Ion Channels/biosynthesis , Ion Channels/genetics , Megakaryocytes/metabolism , Membrane Proteins , Alternative Splicing , Base Sequence , Cell Line , Humans , Megakaryocytes/physiology , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , Sequence Analysis, DNA , TRPC Cation Channels , TRPM Cation Channels , Transcription, Genetic
8.
Thromb Haemost ; 76(6): 1063-71, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8972032

ABSTRACT

Previously cAMP- and cGMP-dependent protein kinases (cAMP-PK, cGMP-PK) have been found predominantly associated with the particulate fraction in human platelets. We now report the distribution and activation of cAMP-PK and cGMP-PK in highly purified fractions of human platelet plasma (PM) and intracellular membranes (IM) prepared using high voltage free flow electrophoresis. Two non-hydrolysable analogues of cAMP and cGMP namely Sp-5,6-DCI-cBiMPS and 8-p-CPT-cGMP have been used to activate cAMP-PK and cGMP-PK respectively. Addition of either agonist with [gamma 32P]ATP stimulated the endogenous activity of cAMP-PK or cGMP-PK in PM but not in IM. With PM Sp-5,6-DCI-cBiMPS stimulated the phosphorylation of protein substrates of Mr 16, 22, 24, 46-50, 66, 90, 160 and 250 kDa. A specific peptide inhibitor of cAMP-PK inhibited the phosphorylation of all of the substrates by Sp-5,6-DCI-cBiMPS. 8-pCPT-cGMP also induced the phosphorylation of a number of substrates particularly 16, 22, 46-50, 90 and 250 kDa proteins. Inclusion of the cAMP-PK inhibitor peptide totally blocked the phosphorylation of the 16 and 22 kDa proteins, partially inhibited phosphorylation of 46-50 and 90 kDa proteins and had no effect on the 250 kDa protein indicating the 46-50, 90 and 250 kDa proteins were also cGMP-PK substrates. Western blotting with antibodies to cGMP-PK and the catalytic subunit of cAMP-PK revealed the presence of the kinases to be exclusively associated with PM with no detection in IM. The presence of cAMP-PK substrates in IM was investigated by exogenous addition of catalytic subunit of cAMP-PK. Phosphoproteins of Mr 16, 22, 27, 30, 45, 75, 116 and 250 kDa were detected. A range of antibodies to cAMP-PK substrates were used to identify and localise the substrates. These antibodies revealed GPIb and VASP to be exclusively associated with PM fractions. Rap IB was also predominantly associated with PM with a small level detected in IM. Antibodies to the IP3 receptor (18A 10 and 4C11) revealed the protein to be predominantly associated with IM. Additionally the antibody 4C11 recognised a 230 kDa protein band in PM that was not seen in IM. From the known specificity of these antibodies the results confirm the presence of a type 1 IP3 receptor in IM and a distinct (possible type III) IP3 receptor with the PM. The 16, 22, 27, 30, 75 and 116 kDa proteins in IM represent newly detected substrates for cAMP-PK of presently unknown identity.


Subject(s)
Blood Platelets/enzymology , Cell Membrane/enzymology , Cyclic AMP-Dependent Protein Kinases/analysis , Cyclic GMP-Dependent Protein Kinases/analysis , Blood Platelets/ultrastructure , Enzyme Activation , Humans , Phosphorylation
9.
J Biol Chem ; 271(1): 289-94, 1996 Jan 05.
Article in English | MEDLINE | ID: mdl-8550575

ABSTRACT

Previous studies have shown that the neurotoxin dendroaspin and the disintegrin kistrin, which show little overall sequence homology but similar residues around RGD (PRGDMP), preferentially inhibited platelet adhesion to fibrinogen. In contrast, the elegantin which has different amino acids around RGD (ARGDNP) preferentially inhibited platelet adhesion to fibronectin. To investigate further the role of amino acids around RGD in disintegrins, we have constructed the genes of a wild-type and of two mutant dendroaspins with substitutions around the RGD, namely [Asn46]- and [Ala42,Asn46]-dendroaspins. Proteins were expressed in Escherichia coli as glutathione S-transferase fusion recombinants and purified to homogeneity by affinity chromatography and reversed phase high performance liquid chromatography. Platelet aggregation studies revealed that wild-type dendroaspin showed an IC50 value similar to that of native dendroaspin, with [Ala42,Asn46]-dendroaspin showing an IC50 value similar to that of elegantin. Interestingly, in platelet adhesion assays, the mutants showed a progressive shift in inhibitory preference, in particular, [Ala42,Asn46]dendroaspin showed nearly identical behavior as elegantin when fibronectin was the immobilized ligand (IC50 = 0.33 microM and 0.6 microM, respectively, compared with 20 microM for native dendroaspin). Native and recombinant wild-type dendroaspin bound to a single class of binding site exhibiting a Kd = 67 nM; [Asn46]- and [Ala42,Asn46]dendroaspins, however, both produced biphasic isotherms with Kd values = 87 nM and 361 nM for [Asn46]dendroaspin and 33 nM and 371 nM for [Ala42,Asn46]dendroaspin, which are close to those of elegantin (Kd values = 18 nM and 179 nM). These studies prove that the amino acids flanking RGD provide an extended locus that regulate the affinity and selectivity of RGD protein dendroaspin.


Subject(s)
Elapid Venoms/genetics , Peptides/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Alanine/chemistry , Alanine/genetics , Alanine/metabolism , Amino Acid Sequence , Asparagine/chemistry , Asparagine/genetics , Asparagine/metabolism , Chromatography, Affinity , Chromatography, High Pressure Liquid , Elapid Venoms/chemistry , Elapid Venoms/metabolism , Escherichia coli/genetics , Methionine/chemistry , Methionine/genetics , Methionine/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligopeptides , Proline/chemistry , Proline/genetics , Proline/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
10.
Biochem J ; 312 ( Pt 1): 223-32, 1995 Nov 15.
Article in English | MEDLINE | ID: mdl-7492316

ABSTRACT

We have previously demonstrated [Lu, Williams, Deadman, Salmon, Kakkar, Wilkinson, Baruch, Authi and Rahman (1994) Biochem. J. 304, 929-936] the preferential antagonism of the interactions of the integrin alpha IIb beta 3 on activated platelets with three immobilized glycoprotein ligands (fibrinogen, fibronectin and von Willebrand factor) by a selected panel of snake-venom RGD (Arg-Gly-Asp)-containing proteins including the disintegrins kistrin and elegantin, and the neurotoxin variant dendroaspin. Kistrin and dendroaspin, although structurally unrelated, contain similar amino acids flanking the tripeptide RGD and behaved as identical antagonists preferentially inhibiting platelet adhesion to immobilized fibrinogen as opposed to fibronectin. In contrast, elegantin, which shares extensive sequence similarity with kistrin but has different amino acids around the tripeptide RGD, preferentially inhibited platelet adhesion to immobilized fibronectin as opposed to fibrinogen. To develop further insights into the mechanisms underlying the preferential antagonism shown by the venom proteins in the adhesion studies, we, in the present study, sought to determine the binding properties of kistrin, elegantin and dendroaspin to the alpha IIb beta 3 complex by radioligand kinetic and competition studies. In direct binding experiments, both kistrin and dendroaspin were observed to bind to a single class of binding site on ADP-activated platelets with apparent equilibrium dissociation constant (Kdapp) values of 42 +/- 2 nM and 21 +/- 6 nM respectively. In competition studies, dendroaspin blocked the binding of 125I-labelled kistrin to ADP-activated platelets in a simple competitive manner, with an apparent equilibrium inhibition constant (Kiapp) of 143 +/- 14 nM, from which an indirect Kdapp = 22 nM for dendroaspin was determined. This result suggests that kistrin and dendroaspin bind to the same site on the integrin alpha IIb beta 3 consistent with their similar inhibitory properties. In contrast, elegantin recognized two classes of binding sites on the alpha IIb beta 3 complex with Kdapp values of 10.5 +/- 0.8 nM and 175 +/- 10 nM, and, unlike dendroaspin, did not inhibit the binding of 125I-labelled kistrin to ADP-activated platelets. However, in reciprocal experiments both kistrin and dendroaspin inhibited the binding of 125I-elegantin to ADP-activated platelets in a non-competitive manner, with Kiapp values of 34 +/- 3 nM and 21 +/- 2 nM respectively. Thus elegantin appears to interact with distinct but interacting sites on the alpha IIb beta 3 complex from the binding site of kistrin and dendroaspin, consistent with its distinctive inhibitory preferences as shown in platelet adhesion studies.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Blood Platelets/metabolism , Oligopeptides/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Snake Venoms/metabolism , Adenosine Diphosphate/pharmacology , Amino Acid Sequence , Binding Sites , Binding, Competitive , Blood Platelets/chemistry , Elapid Venoms/chemistry , Elapid Venoms/metabolism , Elapid Venoms/pharmacology , Glycoproteins/metabolism , Humans , Molecular Sequence Data , Neurotoxins/pharmacology , Oligopeptides/metabolism , Peptides/chemistry , Peptides/metabolism , Peptides/pharmacology , Platelet Activation , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Sequence Homology, Amino Acid , Snake Venoms/chemistry
11.
Biochem J ; 308 ( Pt 1): 15-21, 1995 May 15.
Article in English | MEDLINE | ID: mdl-7755558

ABSTRACT

One of the essential roles for platelets in haemostasis is in the potentiation of blood clotting due to the contribution of anionic phospholipid from the surface of the cells, as an essential cofactor to the proteolytic reactions of coagulation (platelet procoagulant activity). Only a limited number of agonists are known to initiate platelet procoagulant activity. In this study the rate of thrombin formation on the platelet surface was observed to increase in a dose-dependent manner upon treatment of washed platelets with heavy-metal compounds. Unlike the immediate increase observed upon treatment of platelets with calcium ionophore, A23187, the change due to these agents was progressive, approaching a maximum after 10 min. The maximum-fold acceleration of the rate of thrombin formation compared with control platelets was calculated for HgCl2 (56-fold), AgNO3 (42-fold) phenylmercuriacetate (24-fold) and thimerosal (14-fold), compared with 70-fold observed for calcium ionophore. The increase in procoagulant activity due to HgCl2 coincided with a large increase in intracellular calcium and phosphorylation of 22 and 45 kDa proteins. It is considered that the mechanism responsible for the increase in procoagulant activity is exposure of anionic phospholipids. This was detected by a 2-fold increase in the binding of 125I-annexin V upon addition of HgCl2, compared with resting platelets (3-fold on treatment of platelets with calcium ionophore). In contrast to the generation of activity by A23187 and other known agonists of this reaction, heavy-metal compounds appeared to cause little or no release of microparticles from the platelet surface. Since HgCl2 did not cause aggregation of platelets or significant release of serotinin, these findings may give further support to the need for exposure and ligation of glycoprotein IIb:IIIa for vesiculization to occur. Treatment of platelets with heavy metals may constitute a new approach to investigating the early changes in the cell membrane which lead to increased expression of anionic phospholipid.


Subject(s)
Blood Platelets/enzymology , Mercury/pharmacology , Annexin A5/metabolism , Blood Coagulation , Calcium/metabolism , Cell Membrane/ultrastructure , Enzyme Activation/drug effects , Humans , In Vitro Techniques , Phosphoproteins/metabolism , Platelet Aggregation , Prothrombin/metabolism , Serotonin/metabolism
12.
Biochem J ; 306 ( Pt 3): 837-42, 1995 Mar 15.
Article in English | MEDLINE | ID: mdl-7702581

ABSTRACT

The Ca2+ATPase activities of highly purified human platelet membranes prepared by high-voltage free-flow electrophoresis have been analysed by using [gamma-32P]ATP hydrolysis, recognition by antibodies and phosphoenzyme-complex formation. The Ca2+ATPase activity present in mixed membranes was found to be predominantly associated with intracellular membranes after subfractionation, with only a low level of activity associated with plasma membranes. The intracellular-membrane Ca2+ATPase activity was inhibited totally with thapsigargin (Tg), whereas the plasma-membrane Ca2+ATPase was not significantly affected, suggesting that the latter does not belong to the SERCA (sarco-endoplasmic-reticulum Ca2+ATPase) class. A monoclonal antibody, 5F10, raised to the red-cell membrane Ca2+ATPase [Cheng, Magocsi, Cooper, Penniston and Borke (1993) Cell Physiol. Biochem. 4, 31-43] recognized two bands at 135 and 150 kDa in mixed membranes and plasma membranes, and the corresponding bands in red-blood-cell membranes, confirming the Ca2+ATPase to be of the PMCA (plasma-membrane Ca2+ATPase) type. No recognition of any band was detected in intracellular membranes. Identification of the intracellular-membrane Ca2+ATPase activity was carried out with polyclonal antibodies with known specificity towards SERCA 2b (S.2b) and SERCA 3 (N89), and a monoclonal antibody, PL/IM 430, raised against platelet intracellular membranes. All of these antibodies recognized the 100 kDa Ca2+ATPase in mixed membranes and intracellular membranes, with little or no recognition of the activity in the plasma membranes. In some membrane preparations the antibody PL/IM 430 and antiserum N89 recognized similar degradation products, of 74, 70 and 40 kDa, in the intracellular-membrane fraction. The Ca2+ATPase recognized by PL/IM 430 was immunoprecipitated, and the immunoprecipitated protein was specifically recognized by the antiserum N89, but not by S.2b. Analysis of the phosphoenzyme-complex formation revealed potent phosphorylation of the 100 and 74 kDa peptides, both recognized by PL/IM 430 and N89. These studies report the presence of a PMCA in a purified plasma-membrane fraction from human platelets, and that the antibody PL/IM 430 recognizes the SERCA 3 Ca2+ATPase in intracellular membranes.


Subject(s)
Blood Platelets/enzymology , Calcium-Transporting ATPases/analysis , Cell Membrane/enzymology , Humans , Immunoblotting
13.
Biochim Biophys Acta ; 1224(3): 480-8, 1994 Dec 30.
Article in English | MEDLINE | ID: mdl-7803507

ABSTRACT

The relationship between platelet aggregation, calpain activation, PKC activities and the secretory response have been examined in PMA-and ionomycin-stimulated platelets. Co-addition of PMA and ionomycin resulted in a maximal synergistic secretion of [14C]5-hydroxytryptamine ([14C]5-HT) from platelet dense granules. However, prior addition of PMA for 5 or 10 min resulted in a reduction of this secretory response. Inclusion of either RGDS (to inhibit platelet aggregation) or E64-d (to inhibit calpain activity) resulted in full restoration of the secretory response. In experiments to determine the activity status of PKC, PMA was found to induce a loss in cytosolic and total PKC activity without an increase in membrane-associated activities during this time period. Inhibition of either platelet aggregation or calpain activity resulted in preservation of total and cytosolic activities with a measurable increase in membrane translocated activity. PMA-induced phosphorylation of a number of PKC substrates was measured in 32P-labelled platelets. PMA induced potent phosphorylation of the 45 and 20 kDa species and also proteins of the molecular masses 66, 80, 97 and 119 kDa. Phosphorylation was maximal at either 1 or 2 min after which dephosphorylation occurred. Inclusion of either RGDS or E64-d resulted in a reduction of the dephosphorylation rates, and sustained phosphorylation of the 66, 80, 97 and 119 kDa proteins. These studies suggest that the activity status of PKC is an important factor in the level of secretion obtained and that platelet aggregation is involved in calpain-initiated down-regulation of PKC.


Subject(s)
Blood Platelets/drug effects , Calpain/pharmacology , Down-Regulation , Platelet Aggregation/drug effects , Protein Kinase C/metabolism , Blood Platelets/metabolism , Humans , Ionomycin/pharmacology , Male , Oligopeptides/pharmacology , Phosphorylation , Platelet Aggregation Inhibitors/pharmacology , Serotonin/metabolism , Tetradecanoylphorbol Acetate/pharmacology
14.
Biochem J ; 304 ( Pt 3): 929-36, 1994 Dec 15.
Article in English | MEDLINE | ID: mdl-7529494

ABSTRACT

The inhibitory properties of a panel of snake-venom-derived RGD (Arg-Gly-Asp) proteins, including the disintegrins kistrin, elegantin and albolabrin, and the neurotoxin homologue dendroaspin, were investigated in a platelet-adhesion assay using three immobilized ligands of the glycoprotein IIb-IIIa complex (alpha IIb beta 3), namely fibrinogen, fibronectin and von Willebrand factor (vWF). The snake-venom proteins preferentially inhibited the adhesion of ADP-treated platelets to one or more of the immobilized ligands. Kistrin and dendroaspin exhibited similar inhibitory characteristics, abrogating platelet adhesion to fibrinogen and vWF at nanomolar concentrations, but poorly inhibiting adhesion to fibronectin. Kistrin and dendroaspin share little overall amino-acid-sequence identity, but a considerable level of sequence similarity exists around the RGD tripeptide. Synthetic cyclic peptides corresponding to these regions of kistrin and dendroaspin inhibited platelet adhesion to both fibrinogen and fibronectin with approximately equal potency, but were 100-fold weaker antagonists of the interactions of the alpha IIb beta 3 complex with fibrinogen than their parent proteins. The disintegrins elegantin and albolabrin, which share approx. 60% overall amino-acid-sequence similarity with kistrin but have different residues around the RGD tripeptide, exhibited different antagonistic preferences. Elegantin inhibited platelet adhesion to immobilized vWF and fibronectin, but was significantly less effective at disrupting adhesion to fibrinogen. Albolabrin selectively inhibited platelet adhesion to immobilized vWF and was less effective with fibrinogen and fibronectin as adhesive ligands. In contrast with the behaviour of these venom proteins, the adhesion of ADP-treated platelets to immobilized fibrinogen, fibronectin and vWF was inhibited non-selectively by a range of monoclonal antibodies with specificity for the alpha IIb beta 3 complex. These observations, therefore, define antagonistic preferences in this panel of venom proteins towards the interactions of the alpha IIb beta 3 complex with three immobilized glycoprotein ligands.


Subject(s)
Fibrinogen/antagonists & inhibitors , Fibrinogen/metabolism , Fibronectins/antagonists & inhibitors , Fibronectins/metabolism , Integrins/antagonists & inhibitors , Integrins/metabolism , Oligopeptides/pharmacology , Platelet Adhesiveness/drug effects , Snake Venoms/pharmacology , von Willebrand Factor/antagonists & inhibitors , von Willebrand Factor/metabolism , Amino Acid Sequence , Antibodies, Monoclonal , Elapid Venoms/metabolism , Elapid Venoms/pharmacology , Fibrinolytic Agents/metabolism , Fibrinolytic Agents/pharmacology , Humans , Molecular Sequence Data , Oligopeptides/metabolism , Peptides/metabolism , Peptides/pharmacology , Platelet Adhesiveness/physiology , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex , Snake Venoms/metabolism , Structure-Activity Relationship
15.
Biochem Biophys Res Commun ; 202(1): 321-7, 1994 Jul 15.
Article in English | MEDLINE | ID: mdl-8037727

ABSTRACT

Platelets after challenge with alpha-thrombin alone, collagen alone or thrombin/collagen mixture were observed to increase the rate of activation of prothrombin by factor Xa in the presence of factor Va and calcium ion (platelet procoagulant activity) by a maximum of 25, 45 and 110 fold, respectively. The increase in platelet procoagulant activity due to these agonists has been described previously and arises from increased expression of phosphatidylserine on the platelet surface. When platelets were treated with the thrombin receptor activating peptide (TRAP) (SFLLRNPNDKYEPK), alone or in the presence of collagen or thrombin, no change in platelet procoagulant activity was observed at concentrations of TRAP sufficient to cause increased intracellular calcium levels and protein phosphorylation in a manner similar to that of thrombin. In addition, no increase in platelet procoagulant activity was seen upon treatment with TRAP in the presence of inactivated thrombin (PPACK-thrombin). These results suggest that the thrombin-mediated increase in procoagulant activity may be due to activation of a thrombin receptor distinct from the recently cloned G-protein-coupled receptor, or to other proteolytic events on the platelet surface.


Subject(s)
Blood Platelets/physiology , Peptide Fragments/pharmacology , Amino Acid Sequence , Blood Platelets/drug effects , Calcium/blood , Factor Va/metabolism , Factor Xa/metabolism , Fluorescent Dyes , Fura-2/analogs & derivatives , Humans , In Vitro Techniques , Kinetics , Molecular Sequence Data , Prothrombin/metabolism , Receptors, Cell Surface , Thrombin/pharmacology
16.
Biochem J ; 298 Pt 3: 739-42, 1994 Mar 15.
Article in English | MEDLINE | ID: mdl-8141791

ABSTRACT

In the present study we describe the characterization and localization of Ins(1,3,4,5)P4-binding sites in human platelet membranes. Specific binding sites for Ins(1,3,4,5)P4 have been identified on mixed, plasma and intracellular membranes from neuraminidase-treated platelets using highly purified carrier-free [32P]Ins(1,3,4,5)P4. The displacement of Ins(1,3,4,5)P4 from these sites by Ins(1,4,5)P3 and InsP6 occurs at greater than two orders of magnitude higher concentrations and with Ins(1,3,4,5,6)P5 at about 40-fold higher concentrations than with Ins(1,3,4,5)P4. The membranes were further separated by free-flow electrophoresis into plasma and intracellular membranes. The Ins(1,3,4,5)P4-binding sites separated with plasma membranes, and showed similar affinities and specificities as mixed membranes, whereas Ins(1,4,5)P3-binding sites were predominantly in the intracellular membranes. These results suggest a predominantly plasma membrane location for putative Ins(1,3,4,5)P4 receptors in human platelets.


Subject(s)
Blood Platelets/metabolism , Cell Membrane/metabolism , Inositol Phosphates/blood , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Binding Sites , Binding, Competitive , Cattle , Humans , Phosphorus Radioisotopes
17.
J Biol Chem ; 269(2): 1410-6, 1994 Jan 14.
Article in English | MEDLINE | ID: mdl-8288608

ABSTRACT

An organellar-type of Ca2+ pump formerly detected by means of its phosphoprotein intermediate in platelets and in lymphoid cells, and which runs in acid gels at 97 kDa, is now characterized as sarco/endoplasmic reticulum Ca2+ATPase 3 (SERCA3). SERCA3 is co-expressed in these cells along with the housekeeping SERCA2b. This conclusion is based on the following observations. 1) Tryptic digestion the phosphoprotein intermediate of SERCA3 expressed in COS cells yields a phosphorylated fragment of about 80 kDa, which can be clearly distinguished from the 57-kDa fragments formed in the SERCA1 and SERCA2 pumps. This 80-kDa fragment comigrates with a similar phosphoprotein fragment previously observed in human platelets (Papp, B., Enyedi, A., Pászty, K., Kovács, T., Sarkadi, B., Gárdos, G., Wuytack, F., and Enouf, J. (1992) Biochem. J. 288, 297-302). 2) An antiserum directed against an NH2-terminal SERCA3-specific peptide (N89) reacts with SERCA3 expressed in COS cells and with the 97-kDa protein in rat platelets and the corresponding protein in human platelets. Likewise an antiserum against the rat SERCA3 terminus (C90) binds to SERCA3 expressed in COS cells and to the 97-kDa band in rat platelets, but it does not recognize the human platelet pump. In conformity with the predicted absence of the T1 tryptic cleavage site in SERCA3, the autophosphorylated aspartyl residue and the COOH-terminal epitope were co-localized on the 80-kDa fragment. 3) The co-expression of nearly equal levels of SERCA3 and SERCA2b messengers in human lymphoblastoid Jurkat cells and in proliferating rat mucosal mast cells was also demonstrated by reverse transcriptase polymerase chain reaction.


Subject(s)
Blood Platelets/enzymology , Calcium-Transporting ATPases/metabolism , Lymphocytes/enzymology , Mast Cells/enzymology , Amino Acid Sequence , Animals , Base Sequence , Biological Transport, Active , DNA Primers/chemistry , Endoplasmic Reticulum/enzymology , Gene Expression , Humans , Molecular Sequence Data , RNA, Messenger/genetics , Rats , Sarcoplasmic Reticulum/enzymology , Sequence Alignment , Sequence Homology, Amino Acid
18.
Biochem J ; 294 ( Pt 1): 119-26, 1993 Aug 15.
Article in English | MEDLINE | ID: mdl-8363562

ABSTRACT

The effects of the Ca(2+)-ATPase inhibitors thapsigargin (Tg) and 2,5-di-(t-butyl)-1,4-benzohydroquinone (tBuBHQ) were examined by using Ca(2+)-regulatory systems of platelet mixed membranes, saponin-permeabilized and intact platelets. Both agents inhibit Ca(2+)-ATPase activities of platelet mixed membranes, without any effect on the basal Mg(2+)-ATPase activity. Tg is more effective (EC50 = 35 nM) than tBuBHQ (EC50 = 580 nM). The effect of the two inhibitors on 45Ca2+ release from saponin-permeabilized platelets has also been characterized. 45Ca2+ uptake into non-mitochondrial intracellular stores occurs via an ATP-dependent mechanism, and if added at equilibrium the second messenger Ins(1,4,5)P3 releases 50% of the accumulated 45Ca2+. Maximally effective concentrations of Tg (1 microM) and tBuBHQ (50 microM) release 77% and 68% of the accumulated 45Ca2+. Addition of Ins(1,4,5)P3 together with either Tg or tBuBHQ resulted in a non-additive release which was the same as with either Tg or tBuBHQ alone, indicating that the Ins(1,4,5)P3-sensitive Ca2+ pool was a subset of the pool that is sensitive to the Ca(2+)-ATPase inhibitors. Release of 45Ca2+ by either Tg or tBuBHQ was not affected by heparin, which totally blocked Ins(1,4,5)P3-induced Ca2+ release, and Tg was found not to affect [32P]Ins(1,4,5)P3 binding to its receptor on mixed membranes. Thus both Tg and tBuBHQ release Ca2+ from a pool that totally overlaps the Ins(1,4,5)P3-sensitive pool without affecting Ins(1,4,5)P3 function. In intact indomethacin-treated Fura 2-loaded platelets, Tg and tBuBHQ cause Ca2+ elevation, arising from release from intracellular stores and influx from the outside. Both Tg and tBuBHQ elevated Ca2+ to similar levels, which were less and slower than those observed with thrombin. Addition of thrombin to cells already treated with Tg or tBuBHQ produced further elevation of Ca2+, indicating agonist utilization of a Ca(2+)-ATPase inhibitor-insensitive pool. In aggregation experiments Tg and tBuBHQ showed different functional effects. In indomethacin-treated cells Tg induces slow aggregation and secretion responses, whereas tBuBHQ only induces shape change. Both agents show synergistic secretory responses with the protein kinase C activator dioctanoylglycerol (DiC8). Tg also showed greater ability than tBuBHQ to release [3H]arachidonic acid (AA) from [3H]AA-labelled platelets. Additionally, in [32P]Pi-labelled platelets both Tg and tBuBHQ induced phosphorylation of myosin light chain, a 27 kDa protein and the 45 kDa protein pleckstrin, but Tg showed a greater ability than tBuBHQ to cause phosphorylation of pleckstrin. These studies indicate that Tg and tBuBHQ are effective in releasing the Ins(1,4,5)P3-sensitive Ca2+ pool in platelets.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Blood Platelets/drug effects , Calcium/metabolism , Hydroquinones/pharmacology , Platelet Activation , Terpenes/pharmacology , Binding Sites , Blood Platelets/metabolism , Calcium-Transporting ATPases/antagonists & inhibitors , Calcium-Transporting ATPases/metabolism , Cells, Cultured , Humans , In Vitro Techniques , Inositol 1,4,5-Trisphosphate/metabolism , Saponins , Thapsigargin
SELECTION OF CITATIONS
SEARCH DETAIL
...