Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Front Biosci (Landmark Ed) ; 27(5): 161, 2022 05 18.
Article in English | MEDLINE | ID: mdl-35638428

ABSTRACT

BACKGROUND: Platelet-derived extracellular vesicles (PEVs) are small vesicles released by activated platelets that are gaining growing interest in the field of vascular biology. The mode of platelet activation is a critical determinant of PEVs release, phenotype and function. However, only very limited information is available concerning the impact of the platelet purification procedure on PEVs release. METHODS: Washed or isolated platelets were separated by differential centrifugations. For washed platelets, the platelet pellet was washed by resuspension in PIPES buffer and finally resuspended in HEPES buffer. Isolated platelets were obtained by directly resuspending the platelet pellet in HEPES, skipping the washing steps in PIPES buffer. PEVs release was induced in washed or isolated platelets by stimulation with different agonist and analysed by Nanoparticle Tracking Analysis. RESULTS: Isolated platelets showed a higher release of PEVs upon adenosine diphosphate (ADP) stimulation compared to washed platelets, whereas PEVs released upon stimulation with strong agonists (thrombin, collagen, A23187, U46619) were similar in the two groups. This different responsiveness to ADP was also observed as a higher α-granules release and protein kinase C activation in isolated platelets compared to washed ones. Residual plasma contamination appeared to be essential for the ability of platelets to release PEVs in response to ADP. CONCLUSIONS: In conclusion, our study strongly suggests that procedure adopted for platelets preparation is a critical determinant of PEVs release upon ADP stimulation.


Subject(s)
Blood Platelets , Extracellular Vesicles , Adenosine Diphosphate/metabolism , Adenosine Diphosphate/pharmacology , Blood Platelets/metabolism , Extracellular Vesicles/metabolism , HEPES/metabolism , Platelet Activation
2.
Adv Biol Regul ; 59: 36-52, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26159296

ABSTRACT

Blood platelets are anucleated circulating cells that play a critical role in hemostasis and are also implicated in arterial thrombosis, a major cause of death worldwide. The biological function of platelets strongly relies in their reactiveness to a variety of extracellular agonists that regulate their adhesion to extracellular matrix at the site of vascular injury and their ability to form rapidly growing cell aggregates. Among the membrane receptors expressed on the cell surface, integrins are crucial for both platelet activation, adhesion and aggregation. Integrin affinity for specific ligands is regulated by intracellular signaling pathways activated in stimulated platelets, and, once engaged, integrins themselves generate and propagate signals inside the cells to reinforce and consolidate platelet response and thrombus formation. Phosphatidylinositol 3-Kinases (PI3Ks) have emerged as crucial players in platelet activation, and they are directly implicated in the regulation of integrin function. This review will discuss the contribution of PI3Ks in platelet integrin signaling, focusing on the role of specific members of class I PI3Ks and their downstream effector Akt on both integrin inside-out and outside-in signaling. The contribution of the PI3K/Akt pathways stimulated by integrin engagement and platelet activation in thrombus formation and stabilization will also be discussed in order to highlight the possibility to target these enzymes in effective anti-thrombotic therapeutic strategies.


Subject(s)
Blood Platelets/metabolism , Integrins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Thrombosis/metabolism , Animals , Humans , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics
3.
Biochim Biophys Acta ; 1853(8): 1879-88, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25960397

ABSTRACT

Phosphatidylinositol 3-kinaseß (PI3Kß) plays a predominant role in integrin outside-in signaling and in platelet activation by GPVI engagement. We have shown that the tyrosine kinase Pyk2 mediates PI3Kß activation downstream of integrin αIIbß3, and promotes the phosphorylation of the PI3K-associated adaptor protein c-Cbl. In this study, we compared the functional correlation between Pyk2 and PI3Kß upon recruitment of the two main platelet collagen receptors, integrin α2ß1 and GPVI. PI3Kß-mediated phosphorylation of Akt was inhibited in Pyk2-deficient platelets adherent to monomeric collagen through integrin α2ß1, but occurred normally upon GPVI ligation. Integrin α2ß1 engagement led to Pyk2-independent association of c-Cbl with PI3K. However, c-Cbl was not phosphorylated in adherent platelets, and phosphorylation of Akt occurred normally in c-Cbl-deficient platelets, indicating that the c-Cbl is dispensable for Pyk2-mediated PI3Kß activation. Stimulation of platelets with CRP, a selective GPVI ligand, induced c-Cbl phosphorylation in the absence of Pyk2, but failed to promote its association with PI3K. Pyk2 activation was completely abrogated in PI3KßKD, but not in PI3KγKD platelets, and was strongly inhibited by Src kinases and phospholipase C inhibitors, and by BAPTA-AM. The absence of PI3Kß activity also hampered GPVI-induced tyrosine-phosphorylation and activation of PLCγ2, preventing intracellular Ca2+ increase and phosphorylation of pleckstrin. Moreover, GPVI-induced intracellular Ca2+ increase and pleckstrin phosphorylation were also strongly inhibited in human platelets treated with the PI3Kß inhibitor TGX-221. These results outline important differences in the regulation of PI3Kß by GPVI and integrin α2ß1 and suggest that inhibition of Pyk2 may target PI3Kß activation in a selective context of platelet stimulation.


Subject(s)
Focal Adhesion Kinase 2/physiology , Integrin alpha2beta1/physiology , Phosphatidylinositol 3-Kinases/metabolism , Platelet Membrane Glycoproteins/physiology , Proto-Oncogene Proteins c-cbl/physiology , Animals , Cells, Cultured , Enzyme Activation , Humans , Mice , Mice, Knockout , Signal Transduction
4.
Biochem J ; 469(2): 199-210, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25967238

ABSTRACT

In blood platelets, stimulation of G protein-coupled receptors (GPCRs) by thrombin triggers the activation of Src family kinases (SFKs), resulting in the tyrosine-phosphorylation of multiple substrates, but the mechanism underlying this process is still poorly understood. In the present study, we show that the time-dependent protein-tyrosine phosphorylation triggered by thrombin in human or murine platelets was totally suppressed only upon concomitant chelation of intracellular Ca(2+) and inhibition of SFKs. Thrombin-induced activation of SFKs was regulated by intracellular Ca(2+) and accordingly the Ca(2+) ionophore A23187 was sufficient to stimulate SFKs. A23187 also triggered the phosphorylation and activation of the Ca(2+)-dependent focal adhesion kinase Pyk2 and Pyk2 activation by thrombin was Ca(2+)-dependent. Stimulation of SFKs by thrombin or A23187 was strongly reduced in platelets from Pyk2 knockout (KO) mice, as was the overall pattern of protein-tyrosine phosphorylation. By immunoprecipitation experiments, we demonstrate that Lyn and Fyn, but not Src, were activated by Pyk2. Inhibition of SFKs by PP2 also reduced the phosphorylation of Pyk2 in thrombin or A23187-stimulated platelets. Analysis of KO mice demonstrated that Fyn, but not Lyn, was required for complete Pyk2 phosphorylation by thrombin. Finally, PP2 reduced aggregation of murine platelets to a level comparable to that of Pyk2-deficient platelets, but did not have further effects in the absence of Pyk2. These results indicate that in thrombin-stimulated platelets, stimulation of Pyk2 by intracellular Ca(2+) initiates SFK activation, establishing a positive loop that reinforces the Pyk2/SFK axis and allows the subsequent massive tyrosine phosphorylation of multiple substrates required for platelet aggregation.


Subject(s)
Blood Platelets/enzymology , Calcium Signaling/drug effects , Focal Adhesion Kinase 2/metabolism , Hemostatics/pharmacology , Proto-Oncogene Proteins c-fyn/metabolism , Thrombin/pharmacology , src-Family Kinases/metabolism , Animals , Blood Platelets/cytology , Calcium Signaling/physiology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Focal Adhesion Kinase 2/genetics , Humans , Mice , Mice, Knockout , Phosphorylation/drug effects , Phosphorylation/physiology , Platelet Activation/drug effects , Platelet Activation/genetics , Proto-Oncogene Proteins c-fyn/genetics , src-Family Kinases/genetics
5.
Biochem J ; 462(3): 513-23, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24984073

ABSTRACT

Alzheimer's disease is associated with the accumulation of Aß (amyloid ß)-peptides in the brain. Besides their cytotoxic effect on neurons, Aß-peptides are thought to be responsible for the atherothrombotic complications associated with Alzheimer's disease, which are collectively known as cerebrovascular disease. In the present study, we investigated the effect of Aß-peptides on human platelet signal transduction and function. We discovered that the 25-35 domain of Aß-peptides induce an increase in platelet intracellular Ca2+ that stimulates α-granule and dense granule secretion and leads to the release of the secondary agonist ADP. Released ADP acts in an autocrine manner as a stimulant for critical signalling pathways leading to the activation of platelets. This includes the activation of the protein kinases Syk, protein kinase C, Akt and mitogen-activated protein kinases. Ca2+-dependent release of ADP is also the main component of the activation of the small GTPase Rap1b and the fibrinogen receptor integrin αIIbß3, which leads to increased platelet aggregation and increased thrombus formation in human whole blood. Our discoveries complement existing understanding of cerebrovascular dementia and suggest that Aß-peptides can induce vascular complications of Alzheimer's disease by stimulating platelets in an intracellular Ca2+-dependent manner. Despite a marginal ADP-independent component suggested by low levels of signalling activity in the presence of apyrase or P2Y receptor inhibitors, Ca2+-dependent release of ADP by Aß-peptides clearly plays a critical role in platelet activation. Targeting ADP signalling may therefore represent an important strategy to manage the cerebrovascular component of Alzheimer's disease.


Subject(s)
Adenosine Diphosphate/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/physiology , Blood Platelets/metabolism , Calcium/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/physiology , Humans , Peptide Fragments/physiology , Platelet Activation , Platelet Aggregation/drug effects , Signal Transduction
6.
Chem Biol Drug Des ; 83(5): 532-40, 2014 May.
Article in English | MEDLINE | ID: mdl-24666508

ABSTRACT

Boron neutron capture therapy (BNCT) is an anticancer treatment based on the accumulation in the tumor cells of (10) B-containing molecules and subsequent irradiation with low-energy neutrons, which bring about the decay of (10) B to very toxic (7) Li(3+) and (4) He(2+) ions. The effectiveness of BNCT is limited by the low delivery and accumulation of the used (10) B-containing compounds. Here, we report the development of folic acid-conjugated 4-amino-phenylboronate as a novel possible compound for the selective delivery of (10) B in BNCT. An extensive analysis about its biocompatibility to mature blood cells and platelet progenitors revealed that the compound markedly supports platelet aggregation, neutrophil oxidative burst, and inhibition of megakaryocyte development, while it does not have any manifest effect on red blood cells.


Subject(s)
Boronic Acids/chemistry , Folic Acid/chemistry , Neutrophils/drug effects , Blood Platelets/cytology , Blood Platelets/drug effects , Boron Neutron Capture Therapy , Boronic Acids/chemical synthesis , Boronic Acids/pharmacology , Cell Differentiation/drug effects , Humans , Megakaryocytes/cytology , Platelet Aggregation/drug effects
7.
Nanomedicine ; 10(3): 589-97, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24161383

ABSTRACT

Boron neutron capture therapy (BNCT) is a radiotherapy treatment based on the accumulation in the tumor of a (10)B-containing drug and subsequent irradiation with low energy neutrons, which bring about the decay of (10)B to (7)Li and an α particle, causing the death of the neoplastic cell. The effectiveness of BNCT is limited by the low delivery and accumulation of the used boron-containing compounds. Here we report the development and the characterization of BPO4 nanoparticles (NPs) as a novel possible alternative drug for BNCT. An extensive analysis of BPO4 NP biocompatibility was performed using both mature blood cells (erythrocytes, neutrophils and platelets) and a model of hematopoietic progenitor cells. A time- and concentration-dependent cytotoxicity study was performed on neoplastic coloncarcinoma and osteosarcoma cell lines. BPO4 functionalization with folic acid, introduced to improve the uptake by tumor cells, appeared to effectively limit the unwanted effects of NPs on the analyzed blood components. FROM THE CLINICAL EDITOR: Boron neutron capture therapy (BNCT) is a radiotherapy treatment modality based on the accumulation of a (10)B-containing drug and subsequent irradiation with low energy neutrons, inducing the decay of (10)B to (7)Li and an α particle, causing neoplastic cell death. This team of authors reports on a folic acid functionalized BPO4 nanoparticle with improved characteristics compared with conventional BNCT approaches, as demonstrated in tumor cell lines, and hopefully to be followed by translational human studies.


Subject(s)
Boron Compounds/pharmacology , Boron Neutron Capture Therapy , Nanoparticles/chemistry , Neoplasms/radiotherapy , Phosphates/pharmacology , Boron Compounds/chemistry , Boron Compounds/pharmacokinetics , Boron Neutron Capture Therapy/methods , Cell Line, Tumor , Folic Acid/chemistry , Folic Acid/metabolism , Humans , Nanoparticles/metabolism , Phosphates/chemistry , Phosphates/pharmacokinetics
8.
Nanomedicine ; 8(8): 1329-36, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22542822

ABSTRACT

Nanomaterials are attracting growing interest for their potential use in several applications as nanomedicine; therefore, the analysis of their potential toxic effects on various cellular models, including circulating blood cells, is mandatory. This study aimed to investigate the effect of three unrelated nanomaterials, namely nanoscale silica, multiwalled carbon nanotubes, and carbon black, on platelet activation and aggregation. We found that these nanomaterials stimulate some of the typical biochemical pathways involved in canonical platelet activation, such as the stimulation of phospholipase C and Rap1b, resulting in the integrin α(IIb)ß3-mediated platelet aggregation, through a mechanism largely dependent on the release of the extracellular second messengers ADP and thromboxane A2. Importantly, we found that doses of nanoparticles unable to trigger appreciable responses can synergize with subthreshold amounts of physiological agonists to mediate platelet aggregation, indicating that even small amounts of nanomaterials in the bloodstream might contribute to the development of thrombosis. FROM THE CLINICAL EDITOR: In this study, nanosized particles of three virtually unrelated materials (silica, multi-walled carbon nanotubes and carbon black) were investigated regarding their effects on platelet activation and aggregation. All were found to stimulate some of the typical biochemical pathways involved in canonical platelet activation, and were found to have synergistic effects with physiologic platelet activator agonists.


Subject(s)
Nanoparticles , Nanotubes, Carbon , Platelet Activation/drug effects , Soot , Blood Proteins/metabolism , Humans , In Vitro Techniques , Integrin alpha2/blood , Nanoparticles/toxicity , Nanotubes, Carbon/adverse effects , Phosphoproteins/metabolism , Platelet Aggregation/drug effects , Signal Transduction/drug effects , Silicon Dioxide/pharmacology , Soot/adverse effects , Soot/pharmacology , Type C Phospholipases/blood , rap GTP-Binding Proteins/blood
10.
Cell Signal ; 22(11): 1681-7, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20600849

ABSTRACT

Three different surface receptors mediate thrombin-induced activation and aggregation of human blood platelets: the protease activated receptors 1 and 4 (PAR1 and PAR4), and the glycoprotein (GP) Ibalpha of the GPIb-IX-V complex. However, their relative contribution in the stimulation of specific intracellular signaling pathways by thrombin remains largely controversial. In this work, we have shown that activation of PAR1 and PAR4 by thrombin or by selective activating peptides stimulated phospholipase C, tyrosine kinases, as well as the small GTPase Rap1b, promoted actin polymerization and cytoskeleton reorganization. When platelets were desensitized for both PAR1 and PAR4, high doses of thrombin, were unable to activate Rap1b, but produced a still evident stimulation of phospholipase C, as documented by the measurement of intracellular Ca(2+) mobilization and protein kinase C activation. These events were abrogated upon proteolysis of GPIbalpha by the metalloproteinase mocarhagin. In PAR1- and PAR4-desensitized platelets, thrombin also induced tyrosine phosphorylation of some substrates, but, surprisingly, this event was largely independent of GPIbalpha binding, as it persisted upon platelet treatment with mocarhagin. Similarly, thrombin-induced actin polymerization and cytoskeleton reorganization were only minimally altered upon PAR1 and PAR4 inactivation and GPIbalpha proteolysis. Interestingly, none of these events were elicited by enzymatically inactive thrombin. Finally we found that GPIbalpha cleavage reduced, but did not abrogate, platelet aggregation in PAR1- and PAR4-desensitized platelets. These results identify a novel pathway for platelet activation operated by thrombin independently of PAR1, PAR4 and GPIbalpha.


Subject(s)
Platelet Activation/drug effects , Platelet Glycoprotein GPIb-IX Complex/metabolism , Receptor, PAR-1/metabolism , Receptors, Thrombin/metabolism , Thrombin/pharmacology , Actins/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Cytoskeleton/drug effects , Humans , Metalloendopeptidases/pharmacology , Phosphorylation , Platelet Aggregation/drug effects , Type C Phospholipases/metabolism , rap GTP-Binding Proteins/metabolism
11.
J Biol Chem ; 283(43): 28795-805, 2008 Oct 24.
Article in English | MEDLINE | ID: mdl-18755689

ABSTRACT

Stimulation of G(q)-coupled receptors activates phospholipase C and is supposed to promote both intracellular Ca(2+) mobilization and protein kinase C (PKC) activation. We found that ADP-induced phosphorylation of pleckstrin, the main platelet substrate for PKC, was completely inhibited not only by an antagonist of the G(q)-coupled P2Y1 receptor but also upon blockade of the G(i)-coupled P2Y12 receptor. The role of G(i) on PKC regulation required stimulation of phosphatidylinositol 3-kinase rather than inhibition of adenylyl cyclase. P2Y12 antagonists also inhibited pleckstrin phosphorylation, Rap1b activation, and platelet aggregation induced upon G(q) stimulation by the thromboxane A(2) analogue U46619. Importantly, activation of phospholipase C and intracellular Ca(2+) mobilization occurred normally. Phorbol 12-myristate 13-acetate overcame the inhibitory effect of P2Y12 receptor blockade on PKC activation but not on Rap1b activation and platelet aggregation. By contrast, inhibition of diacylglycerol kinase restored both PKC and Rap1b activity and caused platelet aggregation. Stimulation of P2Y12 receptor or direct inhibition of diacylglycerol kinase potentiated the effect of membrane-permeable sn-1,2-dioctanoylglycerol on platelet aggregation and pleckstrin phosphorylation, in association with inhibition of its phosphorylation to phosphatidic acid. These results reveal a novel and unexpected role of the G(i)-coupled P2Y12 receptor in the regulation of diacylglycerol-mediated events in activated platelets.


Subject(s)
Blood Platelets/metabolism , Diglycerides/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Receptors, Purinergic P2/metabolism , Signal Transduction , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adenosine Diphosphate/chemistry , Blood Proteins/chemistry , Blood Proteins/pharmacology , Diacylglycerol Kinase/metabolism , Enzyme Inhibitors/pharmacology , Humans , Phosphoproteins/chemistry , Phosphoproteins/pharmacology , Platelet Aggregation , Platelet Aggregation Inhibitors/pharmacology , Protein Kinase C/metabolism , Receptors, Purinergic P2Y12 , Thromboxane A2/metabolism
12.
Cell Signal ; 20(9): 1662-70, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18582561

ABSTRACT

Rap1b and Rap2b are the only members of the Rap family of GTPases expressed in circulating human platelets. Rap1b is involved in the inside-out activation of integrins, while the role of Rap2b is still poorly understood. In this work, we investigated the localization of Rap proteins to specific microdomains of plasma membrane called lipid rafts, implicated in signal transduction. We found that Rap1b was not associated to lipid rafts in resting platelets, and did not translocate to these microdomains in stimulated cells. By contrast, about 20% of Rap2b constitutively associated to lipid rafts, and this percentage did not increase upon platelet stimulation. Rap2b interaction with lipid rafts also occurred in transfected HEK293T cell. Upon metabolic labelling with [(3)H]palmitate, incorporation of the label into Rap2b was observed. Palmitoylation of Rap2b did not occur when Cys176 or Cys177 were mutated to serine, or when the C-terminal CAAX motif was deleted. Contrary to CAAX deletion, Cys176 and Cys177 substitution did not alter the membrane localization of Rap2b, however, relocation of the mutants within lipid rafts was completely prevented. In intact platelets, disruption of Rap2b interaction with lipid rafts obtained by cholesterol depletion caused a significant inhibition of aggregation. Importantly, agonist-induced activation of Rap2b was concomitantly severely impaired. These results demonstrate that Rap2b, but not the more abundant Rap1b, is associated to lipid rafts in human platelets. This interaction is supported by palmitoylation of Rap2b, and is important for a complete agonist-induced activation of this GTPase.


Subject(s)
Blood Platelets/enzymology , Cysteine/metabolism , Lipoylation , Membrane Microdomains/enzymology , Monomeric GTP-Binding Proteins/metabolism , rap GTP-Binding Proteins/metabolism , Amino Acid Sequence , Blood Platelets/drug effects , Cell Line , Cholesterol/deficiency , Detergents/pharmacology , Enzyme Activation/drug effects , Humans , Lipoylation/drug effects , Membrane Microdomains/drug effects , Molecular Sequence Data , Platelet Activation/drug effects , Protein Transport/drug effects , Subcellular Fractions/metabolism , rap GTP-Binding Proteins/chemistry
13.
Thromb Haemost ; 95(3): 483-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16525577

ABSTRACT

We have recently studied a case series of 46 unrelated patients with inherited thrombocytopenias and identified 18 cases that did not fit any known platelet disorder. In two unrelated families, a mild thrombocytopenia with normal platelet size was transmitted in an autosomal dominant fashion. Bleeding time was prolonged in 5 investigated patients. In all of them, flow cytometry and SDS-PAGE of platelet glycoproteins (GP) showed a reduced content of GPIa, a subunit of the GPIa-IIa complex (also known as integrin alpha 2 beta(1)) that is a major collagen receptor on platelets. All other membrane GPs were within the normal range. GPIa deficiency was associated with severely reduced in vitro platelet adhesion to molecules known to interact selectively with GPIa. In vitro platelet aggregation was normal in all subjects, except for a suboptimal platelet response to fibrillar collagen in two patients. A mild defect of alpha-granules was observed in all affected subjects. No mutation was identified in the genes encoding for GPIa or GPIIa. Since no other similar cases have been reported in the literature, we suggest that an autosomal dominant thrombocytopenia associated with GPIa deficiency and alpha-granule defect represents a new form of inherited thrombocytopenia.


Subject(s)
Integrin alpha2/blood , Thrombocytopenia/blood , Adult , Aged , Bleeding Time , Blood Platelets/drug effects , Blood Platelets/metabolism , Collagen/pharmacology , Female , Genes, Dominant , Hemostasis/genetics , Humans , Male , Pedigree , Phenotype , Platelet Adhesiveness , Platelet Aggregation , Thrombocytopenia/diagnosis , Thrombocytopenia/genetics , Thrombospondin 1/metabolism
14.
Cell Signal ; 18(6): 861-70, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16169188

ABSTRACT

The low affinity receptor for immunoglobulin G, FcgammaRIIA, is expressed in human platelets, mediates heparin-induced thrombocytopenia and participates to platelet activation induced by von Willebrand factor. In this work, we found that stimulation of platelets with agonists acting on G-protein-coupled receptors resulted in the tyrosine phosphorylation of FcgammaRIIA, through a mechanism involving a Src kinase. Treatment of platelets with the blocking monoclonal antibody IV.3 against FcgammaRIIA, but not with control IgG, inhibited platelet aggregation induced by TRAP1, TRAP4, the thromboxane analogue U46619, and low concentrations of thrombin. By contrast, platelet aggregation induced by high doses of thrombin was unaffected by blockade of FcgammaRIIA. We also found that the anti-FcgammaRIIA monoclonal antibody IV.3 inhibited pleckstrin phosphorylation and calcium mobilization induced by low, but not high, concentrations of thrombin. In addition, thrombin- or U46619-induced tyrosine phosphorylation of several substrates typically involved in FcgammaRIIA-mediated signalling, such as Syk and PLCgamma2, was clearly reduced by incubation with anti-FcgammaRIIA antibody IV.3. Upon stimulation with thrombin, FcgammaRIIA relocated in lipid rafts, and thrombin-induced tyrosine phosphorylation of FcgammaRIIA occurred within these membrane domains. Controlled disruption of lipid rafts by depleting membrane cholesterol prevented tyrosine phosphorylation of FcgammaRIIA and impaired platelet aggregation induced by U46619 or by low, but not high, concentrations of thrombin. These results indicate that FcgammaRIIA can be activated in human platelets downstream G-protein-coupled receptors and suggest a novel general mechanism for the reinforcement of platelet activation induced by low concentrations of agonists.


Subject(s)
15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Antigens, CD/physiology , Blood Platelets/metabolism , Platelet Activation , Receptors, IgG/physiology , Thrombin/pharmacology , Antigens, CD/metabolism , Cells, Cultured , Humans , Membrane Microdomains/chemistry , Membrane Microdomains/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, IgG/metabolism , Signal Transduction
15.
Blood ; 107(7): 2728-35, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16357324

ABSTRACT

The involvement of the small GTPase Rap1b in platelet integrin alpha2beta1-dependent outside-in signaling was investigated. Platelet adhesion to 4 different specific ligands for integrin alpha2beta1, monomeric collagen, decorin, and collagen-derived peptides CB8(II) and CB11(II), induced a robust and rapid activation of Rap1b. This process did not require secreted ADP or thromboxane A2 production but was critically regulated by phospholipase C (PLC)-derived second messengers. Both Ca2+ and protein kinase C were found to organize independent but additive pathways for Rap1b activation downstream of integrin-alpha2beta1, which were completely blocked by inhibition of PLC with U73122. Moreover, integrin alpha2beta1 engagement failed to trigger Rap1b activation in murine platelets lacking CalDAG-GEFI, a guanine nucleotide exchange factor regulated by Ca2+ and diacylglycerol, despite normal phosphorylation and activation of PLCgamma2. In addition, CalDAG-GEFI-deficient platelets showed defective integrin alpha2beta1-dependent adhesion and spreading. We found that outside-in signaling through integrin alpha2beta1 triggered inside-out activation of integrin alphaIIbbeta3 and promoted fibrinogen binding. Similarly to Rap1b stimulation, this process occurred downstream of PLC activation and was dramatically impaired in murine platelets lacking the Rap1 exchange factor CalDAG-GEFI. These results demonstrate that Rap1b is an important element in integrin-dependent outside-in signaling during platelet adhesion and regulates the cross talk between adhesive receptors.


Subject(s)
Integrin alpha2beta1/physiology , Platelet Adhesiveness/physiology , Platelet Glycoprotein GPIIb-IIIa Complex/physiology , Receptor Cross-Talk/physiology , rap GTP-Binding Proteins/metabolism , Enzyme Activation , Fibrinogen/metabolism , Humans , Integrin alpha2beta1/blood , Kinetics , Second Messenger Systems/physiology , Type C Phospholipases/blood , rap GTP-Binding Proteins/blood
16.
J Pharm Biomed Anal ; 41(1): 36-42, 2006 Apr 11.
Article in English | MEDLINE | ID: mdl-16293388

ABSTRACT

The sulphation pattern of glycosaminoglycan (GAG) plays a critical role in biological functions of proteoglycans. In this study, we showed that decorins from different bovine tissues present specific sulphation pattern coupled with peculiar biological activity. In order to elucidate chemical structure of decorin glycosaminoglycan chains, we improved an electrophoretic method to analyse fluorescent disaccharides from dermatan/chondroitin sulphate GAG chains. The disaccharide separation is based on minigels, and this technique was able to define the polysaccharide chain composition in terms of sulphated and not sulphated disaccharides. This approach allowed not only the measurement of few picomoles of material, but it also permits a rapid qualitative analysis of the GAG chains. Data obtained by PAGEFS indicate that the sulphation pattern of GAG is tissue specific and this finding may explain the different binding properties to von Willebrand factor of decorins.


Subject(s)
Electrophoresis, Polyacrylamide Gel/methods , Extracellular Matrix Proteins/analysis , Extracellular Matrix Proteins/isolation & purification , Glycosaminoglycans/chemistry , Proteoglycans/analysis , Proteoglycans/isolation & purification , Animals , Cattle , Chondroitin Sulfates/chemistry , Chromatography, High Pressure Liquid , Decorin , Dermatan Sulfate/chemistry , Electrophoresis , Extracellular Matrix Proteins/chemistry , Models, Chemical , Polysaccharides/chemistry , Protein Binding , Proteoglycans/chemistry
17.
FEBS Lett ; 574(1-3): 95-100, 2004 Sep 10.
Article in English | MEDLINE | ID: mdl-15358546

ABSTRACT

The small proteoglycan decorin plays an important role in the organisation of the extracellular matrix by binding to several components, including collagen and fibronectin. In this work, we report the dose-dependent and saturable interaction of decorin with the adhesive glycoprotein, von Willebrand factor (VWF). This interaction was mediated by the glycosaminoglycan side chain of decorin and was critically regulated by the degree of sulfation, but not by the amount of iduronic acid. Both chondroitin sulfate and dermatan sulfate, in addition to heparin, were found to bind VWF equally well. Although soluble decorin prevented VWF binding to heparin, purified VWF-A1 domain failed to interact with the proteoglycan. These results identify VWF as a new partner for the small proteoglycan, decorin, in the structural organisation of the extracellular matrix.


Subject(s)
Proteoglycans/metabolism , von Willebrand Factor/metabolism , Decorin , Extracellular Matrix Proteins , Protein Binding , Proteoglycans/chemistry
18.
Biochim Biophys Acta ; 1640(1): 43-51, 2003 Apr 07.
Article in English | MEDLINE | ID: mdl-12676353

ABSTRACT

Adhesion of blood platelets to fibrillar collagens plays a crucial role in haemostasis. Collagen type II is a homotrimeric member of the fibrillar collagen family, whose ability to interact with platelets has been poorly investigated. In this work, we analysed platelet adhesion to the whole collagen type II molecule, as well as to its CNBr peptides. We found that collagen type II is as efficient as collagen type I in supporting platelet adhesion. Platelet binding sites on collagen type II were identified in two different CNBr-derived peptides, CB8 and CB11. The ability of these peptides to support platelet adhesion required the triple helical conformation. Interaction of platelets with CB8 and CB11 peptides was totally dependent on the presence of Mg(2+) ions, and was completely inhibited by the anti-integrin alpha(2)beta(1) antibody P1E6. Upon adhesion to CB8 and CB11, a significant increase in intracellular protein tyrosine phosphorylation was observed. The pattern of tyrosine phosphorylated proteins in CB8- and CB11-adherent platelets was very similar to that observed in platelets adherent to the whole collagen molecule. By immunoprecipitation experiments, we identified two substrates that were tyrosine phosphorylated in adherent platelets as the tyrosine kinase Syk and the PLCgamma2 isozyme. By contrast, platelet adhesion to CB8 and CB11 did not promote tyrosine phosphorylation of FcR gamma-chain. Finally, we found that collagen type II, but not the CNBr-derived peptides, was able to induce cell aggregation associated to protein tyrosine phosphorylation when added to a platelet suspension. These results identify the CNBr peptides from collagen type II CB8 and CB11 as ligands for platelet integrin alpha(2)beta(1), and recognise their ability to support platelet adhesion and activation.


Subject(s)
Blood Platelets/metabolism , Collagen Type II/metabolism , Integrin alpha2beta1/metabolism , Peptides/metabolism , Cell Adhesion , Collagen/metabolism , Cyanogen Bromide/chemistry , Humans , Magnesium/metabolism , Peptide Fragments/metabolism , Peptides/chemistry , Platelet Activation , Protein-Tyrosine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...