Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Arterioscler Thromb Vasc Biol ; 40(10): 2391-2403, 2020 10.
Article in English | MEDLINE | ID: mdl-32787521

ABSTRACT

OBJECTIVE: Reelin, a secreted glycoprotein, was originally identified in the central nervous system, where it plays an important role in brain development and maintenance. In the cardiovascular system, reelin plays a role in atherosclerosis by enhancing vascular inflammation and in arterial thrombosis by promoting platelet adhesion, activation, and thrombus formation via APP (amyloid precursor protein) and GP (glycoprotein) Ib. However, the role of reelin in hemostasis and arterial thrombosis is not fully understood to date. Approach and Results: In the present study, we analyzed the importance of reelin for cytoskeletal reorganization of platelets and thrombus formation in more detail. Platelets release reelin to amplify alphaIIb beta3 integrin outside-in signaling by promoting platelet adhesion, cytoskeletal reorganization, and clot retraction via activation of Rho GTPases RAC1 (Ras-related C3 botulinum toxin substrate) and RhoA (Ras homolog family member A). Reelin interacts with the collagen receptor GP (glycoprotein) VI with subnanomolar affinity, induces tyrosine phosphorylation in a GPVI-dependent manner, and supports platelet binding to collagen and GPVI-dependent RAC1 activation, PLC gamma 2 (1-phosphatidylinositol-4,5-bisphosphate phosphodiesterase gamma-2) phosphorylation, platelet activation, and aggregation. When GPVI was deleted from the platelet surface by antibody treatment in reelin-deficient mice, thrombus formation was completely abolished after injury of the carotid artery while being only reduced in either GPVI-depleted or reelin-deficient mice. CONCLUSIONS: Our study identified a novel signaling pathway that involves reelin-induced GPVI activation and alphaIIb beta3 integrin outside-in signaling in platelets. Loss of both, GPVI and reelin, completely prevents stable arterial thrombus formation in vivo suggesting that inhibiting reelin-platelet-interaction might represent a novel strategy to avoid arterial thrombosis in cardiovascular disease.


Subject(s)
Blood Platelets/enzymology , Carotid Artery Injuries/enzymology , Cell Adhesion Molecules, Neuronal/blood , Extracellular Matrix Proteins/blood , Nerve Tissue Proteins/blood , Neuropeptides/blood , Phospholipase C gamma/blood , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Membrane Glycoproteins/metabolism , Serine Endopeptidases/blood , Thrombosis/enzymology , rac1 GTP-Binding Protein/blood , rhoA GTP-Binding Protein/blood , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Blood Coagulation , Carotid Artery Injuries/blood , Carotid Artery Injuries/etiology , Cell Adhesion Molecules, Neuronal/deficiency , Cell Adhesion Molecules, Neuronal/genetics , Clot Retraction , Cytoskeleton/enzymology , Disease Models, Animal , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/genetics , Mice, 129 Strain , Mice, Inbred C3H , Mice, Inbred C57BL , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Platelet Activation , Reelin Protein , Serine Endopeptidases/deficiency , Serine Endopeptidases/genetics , Signal Transduction , Thrombosis/blood , Thrombosis/etiology
2.
J Thromb Haemost ; 17(9): 1500-1510, 2019 09.
Article in English | MEDLINE | ID: mdl-31145836

ABSTRACT

OBJECTIVE: Clinical and laboratory studies have demonstrated that platelets become hyperactive and prothrombotic in conditions of inflammation. We have previously shown that the proinflammatory cytokine interleukin (IL)-6 forms a complex with soluble IL-6 receptor α (sIL-6Rα) to prime platelets for activation by subthreshold concentrations of collagen. Upon being stimulated with collagen, the transcription factor signal transducer and activator of transcription (STAT) 3 in platelets is phosphorylated and dimerized to act as a protein scaffold to facilitate the catalytic action between the kinase Syk and the substrate phospholipase Cγ2 (PLCγ2) in collagen-induced signaling. However, it remains unknown how collagen induces phosphorylation and dimerization of STAT3. METHODS AND RESULTS: We conducted complementary in vitro experiments to show that the IL-6 receptor subunit glycoprotein 130 (GP130) was in physical proximity to the collagen receptor glycoprotein VI (GPVI in membrane lipid rafts of platelets. This proximity allows collagen to induce STAT3 activation and dimerization, and the IL-6-sIL-6Rα complex to activate the kinase Syk and the substrate PLCγ2 in the GPVI signal pathway, resulting in an enhanced platelet response to collagen. Disrupting lipid rafts or blocking GP130-Janus tyrosine kinase (JAK)-STAT3 signaling abolished the cross-activation and reduced platelet reactivity to collagen. CONCLUSION: These results demonstrate cross-talk between collagen and IL-6 signal pathways. This cross-talk could potentially provide a novel mechanism for inflammation-induced platelet hyperactivity, so the IL-6-GP130-JAK-STAT3 pathway has been identified as a potential target to block this hyperactivity.


Subject(s)
Blood Platelets/metabolism , Cytokine Receptor gp130/blood , Membrane Microdomains/physiology , Platelet Membrane Glycoproteins/physiology , Blood Coagulation/drug effects , Collagen/pharmacology , Cytokine Receptor gp130/chemistry , Hemorheology , Humans , Immunoprecipitation , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/blood , Phospholipase C gamma/blood , Phosphorylation , Platelet Aggregation/drug effects , Platelet Aggregation/physiology , Platelet Membrane Glycoproteins/chemistry , Protein Interaction Mapping , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational , STAT3 Transcription Factor/blood
3.
Atherosclerosis ; 281: 62-70, 2019 02.
Article in English | MEDLINE | ID: mdl-30658193

ABSTRACT

BACKGROUND AND AIMS: Platelets play a fundamental role in the increased atherothrombotic risk related to central obesity since they show hyperactivation and lower sensitivity to antiplatelet therapy in obese patients. The main goal of this study was to identify platelet biomarkers related to the risk of atherothrombosis in obese patients, confirm platelet activation levels in these patients, and identify altered activation pathways. METHODS: Platelets were obtained from cohorts of obese patients and age- and sex-matched lean controls. Biochemical and proteome analyses were done by two-dimensional differential in-gel electrophoresis (2D-DIGE), mass spectrometry, and immunoblotting. Functional and mechanistic studies were conducted with aggregation assays and flow cytometry. RESULTS: We confirmed an up-regulation of αIIb and fibrinogen isoforms in platelets from obese patients. A complementary platelet aggregation approach showed platelets from obese patients are hyper-reactive in response to collagen and collagen-related peptide (CRP), revealing the collagen receptor Glycoprotein VI (GPVI) signalling as one of the altered pathways. We also found the active form of Src (pTyr418) is up-regulated in platelets from obese individuals, which links proteomics to aggregation data. Moreover, we showed that CRP-activated platelets present higher levels of tyrosine phosphorylated PLCγ2 in obese patients, confirming alterations in GPVI signalling. In line with the above, flow cytometry studies show higher surface expression levels of total GPVI and GPVI-dimer in obese platelets, both correlating with BMI. CONCLUSIONS: Our results suggest a higher activation state of SFKs-mediated signalling pathways in platelets from obese patients, with a primary involvement of GPVI signalling.


Subject(s)
Blood Platelets/metabolism , Obesity/blood , Platelet Activation , Platelet Membrane Glycoproteins/metabolism , Adolescent , Adult , Body Mass Index , Case-Control Studies , Female , Humans , Male , Obesity/diagnosis , Phospholipase C gamma/blood , Phosphorylation , Platelet Aggregation , Signal Transduction , Up-Regulation , Young Adult
4.
Arterioscler Thromb Vasc Biol ; 38(2): 344-352, 2018 02.
Article in English | MEDLINE | ID: mdl-29146750

ABSTRACT

OBJECTIVE: TRPM7 (transient receptor potential cation channel, subfamily M, member 7) is a ubiquitously expressed bifunctional protein comprising a transient receptor potential channel segment linked to a cytosolic α-type serine/threonine protein kinase domain. TRPM7 forms a constitutively active Mg2+ and Ca2+ permeable channel, which regulates diverse cellular processes in both healthy and diseased conditions, but the physiological role of TRPM7 kinase remains largely unknown. APPROACH AND RESULTS: Here we show that point mutation in TRPM7 kinase domain deleting the kinase activity in mice (Trpm7R/R ) causes a marked signaling defect in platelets. Trpm7R/R platelets showed an impaired PIP2 (phosphatidylinositol-4,5-bisphosphate) metabolism and consequently reduced Ca2+ mobilization in response to stimulation of the major platelet receptors GPVI (glycoprotein VI), CLEC-2 (C-type lectin-like receptor), and PAR (protease-activated receptor). Altered phosphorylation of Syk (spleen tyrosine kinase) and phospholipase C γ2 and ß3 accounted for these global platelet activation defects. In addition, direct activation of STIM1 (stromal interaction molecule 1) with thapsigargin revealed a defective store-operated Ca2+ entry mechanism in the mutant platelets. These defects translated into an impaired platelet aggregate formation under flow and protection of the mice from arterial thrombosis and ischemic stroke in vivo. CONCLUSIONS: Our results identify TRPM7 kinase as a key modulator of phospholipase C signaling and store-operated Ca2+ entry in platelets. The protection of Trpm7R/R mice from acute ischemic disease without developing intracranial hemorrhage indicates that TRPM7 kinase might be a promising antithrombotic target.


Subject(s)
Arterial Occlusive Diseases/blood , Blood Platelets/metabolism , Calcium Signaling , Calcium/blood , Infarction, Middle Cerebral Artery/blood , TRPM Cation Channels/blood , Thrombosis/blood , Animals , Arterial Occlusive Diseases/genetics , Arterial Occlusive Diseases/pathology , Disease Models, Animal , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Lectins, C-Type/blood , Mice, Mutant Strains , Phosphatidylinositol 4,5-Diphosphate/blood , Phospholipase C beta/blood , Phospholipase C gamma/blood , Phosphorylation , Platelet Membrane Glycoproteins/metabolism , Point Mutation , Receptors, Proteinase-Activated/blood , Stromal Interaction Molecule 1/blood , Synaptophysin/blood , TRPM Cation Channels/deficiency , TRPM Cation Channels/genetics , Thrombosis/genetics , Thrombosis/pathology
5.
J Thromb Haemost ; 15(11): 2230-2244, 2017 11.
Article in English | MEDLINE | ID: mdl-28815933

ABSTRACT

Essentials FcγRIIa-mediated thrombocytopenia is associated with drug-dependent antibodies (DDAbs). We investigated the correlation between αIIb ß3 binding epitopes and induction of DDAbs. An FcγRIIa-transgenic mouse model was used to evaluate thrombocytopenia among anti-thrombotics. An antithrombotic with binding motif toward αIIb ß-propeller domain has less bleeding tendency. SUMMARY: Background Thrombocytopenia, a common side effect of Arg-Gly-Asp-mimetic antiplatelet drugs, is associated with drug-dependent antibodies (DDAbs) that recognize conformation-altered integrin αIIb ß3 . Objective To explore the correlation between αIIb ß3 binding epitopes and induction of DDAb binding to conformation-altered αIIb ß3 , we examined whether two purified disintegrins, TMV-2 and TMV-7, with distinct binding motifs have different effects on induction of αIIb ß3 conformational change and platelet aggregation in the presence of AP2, an IgG1 inhibitory mAb raised against αIIb ß3 . Methods We investigated the possible mechanisms of intrinsic platelet activation of TMV-2 and TMV-7 in the presence of AP2 by examining the signal cascade, tail bleeding time and immune thrombocytopenia in Fc receptor γ-chain IIa (FcγRIIa) transgenic mice. Results TMV-7 has a binding motif that recognizes the αIIb ß-propeller domain of αIIb ß3 , unlike that of TMV-2. TMV-7 neither primed the platelets to bind ligand, nor caused a conformational change of αIIb ß3 as identified with the ligand-induced binding site mAb AP5. In contrast to eptifibatide and TMV-2, cotreatment of TMV-7 with AP2 did not induce FcγRIIa-mediated platelet aggregation and the downstream activation cascade. Both TMV-2 and TMV-7 efficaciously prevented occlusive thrombosis in vivo. Notably, both eptifibatide and TMV-2 caused severe thrombocytopenia mediated by FcγRIIa, prolonged tail bleeding time in vivo, and repressed human whole blood coagulation indexes, whereas TMV-7 did not impair hemostatic capacity. Conclusions TMV-7 shows antiplatelet and antithrombotic activities resulting from a mechanism different from that of all other tested αIIb ß3 antagonists, and may offer advantages as a therapeutic agent with a better safety profile.


Subject(s)
Antibodies/blood , Blood Platelets/drug effects , Fibrinolytic Agents/pharmacology , Peptides/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Receptors, IgG/metabolism , Thrombocytopenia/chemically induced , Thrombosis/prevention & control , Animals , Antibodies, Monoclonal/pharmacology , Binding Sites , Blood Platelets/metabolism , Disease Models, Animal , Eptifibatide , Fibrinolytic Agents/immunology , Fibrinolytic Agents/toxicity , Genetic Predisposition to Disease , Humans , Immunoreceptor Tyrosine-Based Activation Motif , Male , Mice, Inbred ICR , Mice, Transgenic , Peptides/immunology , Peptides/toxicity , Phenotype , Phospholipase C gamma/blood , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/immunology , Platelet Aggregation Inhibitors/toxicity , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Platelet Glycoprotein GPIIb-IIIa Complex/immunology , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Conformation , Receptors, IgG/genetics , Structure-Activity Relationship , Syk Kinase/blood , Thrombocytopenia/blood , Thrombocytopenia/immunology , Thrombosis/blood , Thrombosis/genetics
6.
Arterioscler Thromb Vasc Biol ; 36(8): 1507-16, 2016 08.
Article in English | MEDLINE | ID: mdl-27339458

ABSTRACT

OBJECTIVE: Activation of platelets by subendothelial collagen results in an increase of cytosolic Ca(2+) concentration ([Ca(2+)]i) and is followed by platelet activation and thrombus formation that may lead to vascular occlusion. The present study determined the role of phosphoinositide-dependent protein kinase 1 (PDK1) in collagen-dependent platelet Ca(2+) signaling and ischemic stroke in vivo. APPROACH AND RESULTS: Platelet activation with collagen receptor glycoprotein VI agonists collagen-related peptide or convulxin resulted in a significant increase in PDK1 activity independent of second-wave signaling. PDK1 deficiency was associated with reduced platelet phospholipase Cγ2-dependent inositol-1,4,5-trisphosphate production and intracellular [Ca(2+)]i in response to stimulation with collagen-related peptide or convulxin. The defective increase of [Ca(2+)]i resulted in a substantial defect in activation-dependent platelet secretion and aggregation on collagen-related peptide stimulation. Furthermore, Rac1 activation and spreading, adhesion to collagen, and thrombus formation under high arterial shear rates were significantly diminished in PDK1-deficient platelets. Mice with PDK1-deficient platelets were protected against arterial thrombotic occlusion after FeCl3-induced mesenteric arterioles injury and ischemic stroke in vivo. These mice had significantly reduced brain infarct volumes, with a significantly increased survival of 7 days after transient middle cerebral artery occlusion without increase of intracerebral hemorrhage. Tail bleeding time was prolonged in pdk1(-/-) mice, reflecting an important role of PDK1 in primary hemostasis. CONCLUSIONS: PDK1 is required for Ca(2+)-dependent platelet activation on stimulation of collagen receptor glycoprotein VI, arterial thrombotic occlusion, and ischemic stroke in vivo.


Subject(s)
3-Phosphoinositide-Dependent Protein Kinases/metabolism , Blood Platelets/enzymology , Calcium Signaling , Collagen/metabolism , Infarction, Middle Cerebral Artery/enzymology , Platelet Activation , Thrombosis/enzymology , 3-Phosphoinositide-Dependent Protein Kinases/deficiency , 3-Phosphoinositide-Dependent Protein Kinases/genetics , Animals , Disease Models, Animal , Genetic Predisposition to Disease , Infarction, Middle Cerebral Artery/blood , Infarction, Middle Cerebral Artery/pathology , Inositol 1,4,5-Trisphosphate/blood , Mice, Knockout , Neuropeptides/blood , Phenotype , Phospholipase C gamma/blood , Platelet Adhesiveness , Platelet Aggregation , Platelet Membrane Glycoproteins/deficiency , Platelet Membrane Glycoproteins/genetics , Thrombosis/blood , Thrombosis/pathology , Time Factors , rac1 GTP-Binding Protein/blood
7.
Arterioscler Thromb Vasc Biol ; 36(5): 846-54, 2016 05.
Article in English | MEDLINE | ID: mdl-26988594

ABSTRACT

OBJECTIVE: Reactive oxygen species (ROS) are known to regulate platelet activation; however, the mechanisms of ROS production during platelet activation remain unclear. Platelets express different isoforms of nicotinamide adenine dinucleotide (phosphate) (NAD(P)H) oxidases (NOXs). Here, we investigated the role of NOX1 and NOX2 in ROS generation and platelet activation using NOX1 and NOX2 knockout mice. APPROACH AND RESULTS: NOX1(-/Y) platelets showed selective defects in G-protein-coupled receptor-mediated platelet activation induced by thrombin and thromboxane A2 analog U46619, but were not affected in platelet activation induced by collagen-related peptide, a glycoprotein VI agonist. In contrast, NOX2(-/-) platelets showed potent inhibition of collagen-related peptide-induced platelet activation, and also showed partial inhibition of thrombin-induced platelet activation. Consistently, production of ROS was inhibited in NOX1(-/Y) platelets stimulated with thrombin, but not collagen-related peptide, whereas NOX2(-/-) platelets showed reduced ROS generation induced by collagen-related peptide or thrombin. Reduced ROS generation in NOX1/2-deficient platelets is associated with impaired activation of Syk and phospholipase Cγ2, but minimally affected mitogen-activated protein kinase pathways. Interestingly, laser-induced arterial thrombosis was impaired but the bleeding time was not affected in NOX2(-/-) mice. Wild-type thrombocytopenic mice injected with NOX2(-/-) platelets also showed defective arterial thrombosis, suggesting an important role for platelet NOX2 in thrombosis in vivo but not hemostasis. CONCLUSIONS: NOX1 and NOX2 play differential roles in different platelet activation pathways and in thrombosis. ROS generated by these enzymes promotes platelet activation via the Syk/phospholipase Cγ2/calcium signaling pathway.


Subject(s)
Blood Platelets/enzymology , Membrane Glycoproteins/blood , NADH, NADPH Oxidoreductases/blood , NADPH Oxidases/blood , Platelet Activation , Reactive Oxygen Species/blood , Thrombosis/blood , Thrombosis/enzymology , Animals , Blood Platelets/drug effects , CD11b Antigen/blood , Calcium Signaling , Disease Models, Animal , Enzyme Activation , Genetic Predisposition to Disease , Hemostasis , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , NADH, NADPH Oxidoreductases/deficiency , NADH, NADPH Oxidoreductases/genetics , NADPH Oxidase 1 , NADPH Oxidase 2 , NADPH Oxidases/deficiency , NADPH Oxidases/genetics , Phenotype , Phospholipase C gamma/blood , Phosphorylation , Platelet Activation/drug effects , Platelet Aggregation , Platelet Membrane Glycoproteins/agonists , Platelet Membrane Glycoproteins/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/blood , Syk Kinase/blood , Thrombin/metabolism , Thrombosis/genetics , Time Factors
8.
Thromb Haemost ; 115(5): 969-78, 2016 05 02.
Article in English | MEDLINE | ID: mdl-26790499

ABSTRACT

Adaptor proteins play a critical role in the assembly of signalling complexes after engagement of platelet receptors by agonists such as collagen, ADP and thrombin. Recently, using proteomics, the Dok (downstream of tyrosine kinase) adapter proteins were identified in human and mouse platelets. In vitro studies suggest that Dok-1 binds to platelet integrin ß3, but the underlying effects of Dok-1 on αIIbß3 signalling, platelet activation and thrombosis remain to be elucidated. In the present study, using Dok-1-deficient (Dok-1-/-) mice, we determined the phenotypic role of Dok-1 in αIIbß3 signalling. We found that platelets from Dok-1-/- mice displayed normal aggregation, activation of αIIbß3 (assessed by binding of JON/A), P-selectin surface expression (assessed by anti-CD62P), and soluble fibrinogen binding. These findings indicate that Dok-1 does not affect "inside-out" platelet signalling. Compared with platelets from wild-type (WT) mice, platelets from Dok-1-/- mice exhibited increased clot retraction (p < 0.05 vs WT), increased PLCγ2 phosphorylation, and enhanced spreading on fibrinogen after thrombin stimulation (p < 0.01 vs WT), demonstrating that Dok-1 negatively regulates αIIbß3 "outside-in" signalling. Finally, we found that Dok-1-/- mice exhibited significantly shortened bleeding times and accelerated carotid artery thrombosis in response to photochemical injury (p < 0.05 vs WT mice). We conclude that Dok-1 modulates thrombosis and haemostasis by negatively regulating αIIbß3 outside-in signalling.


Subject(s)
DNA-Binding Proteins/blood , Phosphoproteins/blood , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , RNA-Binding Proteins/blood , Thrombosis/prevention & control , Animals , Bleeding Time , Carotid Artery Thrombosis/blood , Carotid Artery Thrombosis/genetics , Carotid Artery Thrombosis/prevention & control , Clot Retraction , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Fibrinogen/metabolism , Hemostasis , Humans , Mice , Mice, 129 Strain , Mice, Knockout , P-Selectin/blood , Phospholipase C gamma/blood , Phosphoproteins/deficiency , Phosphoproteins/genetics , Platelet Activation , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , RNA-Binding Proteins/genetics , Signal Transduction , Thrombosis/blood , Thrombosis/genetics
9.
Int J Oral Maxillofac Surg ; 43(12): 1418-26, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25085076

ABSTRACT

The aim of this study was to investigate the prognostic and predictive values of phospholipase C gamma 1 (PLCG1) expression in patients with locally advanced and resectable oral squamous cell carcinoma (OSCC), who were treated in a prospective, randomized, phase 3 trial evaluating standard treatment with surgery and postoperative radiation preceded or not by induction docetaxel, cisplatin, and 5-fluorouracil (TPF). Immunohistochemical staining for PLCG1 was performed on the biopsies of 232 out of 256 OSCC patients at clinical stage III/IVA; the PLCG1 positive score was determined by immunoreactive scoring system. The survival analysis was performed by Kaplan-Meier method; hazard ratios were calculated using the Cox proportional hazards model. Patients with a low PLCG1 expression had a significantly better overall survival (P=0.022), and a trend towards better disease-free survival (P=0.087), loco-regional recurrence-free survival (P=0.058), distant metastasis-free survival (P=0.053), and a high response rate to TPF induction chemotherapy with regard to clinical response (P=0.052) and pathological response (P=0.061), compared to those with high PLCG1 expression. Our results suggest that PLCG1 expression could be used as a prognostic biomarker for patients with advanced OSCC; however, it was not an adequate predictive biomarker for TPF induction chemotherapy.


Subject(s)
Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/therapy , Mouth Neoplasms/pathology , Mouth Neoplasms/therapy , Phospholipase C gamma/blood , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers/blood , Biopsy , Carcinoma, Squamous Cell/blood , Combined Modality Therapy , Female , Humans , Immunohistochemistry , Male , Middle Aged , Mouth Neoplasms/blood , Predictive Value of Tests , Prognosis , Prospective Studies , Survival Analysis
10.
Thromb Res ; 129(1): 36-42, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21875743

ABSTRACT

BACKGROUND: Tamoxifen is a selective estrogen receptor antagonist that is widely used for treatment and prevention of breast cancer. However, tamoxifen use can lead to an increased incidence of thrombotic events. The reason for this adverse event remains unknown. Previous studies showed that tamoxifen and its active metabolite Z-4-hydroxytamoxifen rapidly increased intracellular free calcium ([Ca(2+)](i)) in human platelets by a non-genomic mechanism that involved the activation of phospholipase C. Platelets play a pivotal role in thrombosis and Ca(2+) elevation is a central event in platelet activation. Therefore the mechanism by which tamoxifen activated Ca(2+) entry into platelets was investigated. METHODS: [Ca(2+)](i) was measured using the fluorescent indicator fura-2 and reactive oxygen species were measured using lucigenin in isolated human platelets. RESULTS: Tamoxifen analogs E-4-hydroxytamoxifen, with weak activity at the nuclear estrogen receptor and Z-4-hydroxytamoxifen, with strong activity at nuclear estrogen receptor, were equally active at increasing [Ca(2+)](i) and synergizing with ADP and thrombin to increase [Ca(2+)](i) in platelets. This result suggests that the effects of tamoxifen and E- and Z-4-hydroxytamoxifen to increase [Ca(2+)](i) are not mediated by the classical genomic estrogen receptor. The effects of tamoxifen to increase [Ca(2+)](i) were strongly inhibited by apocynin and apocynin dimer. This suggests that tamoxifen activates NADPH oxidase which leads to superoxide generation and in turn caused an increase in [Ca(2+)](i). Free radical scavengers TEMPO and TEMPOL also inhibited tamoxifen-induced [Ca(2+)](i) elevation. Inhibition of phosphoinositide-3-kinase (PI3-kinase), an upstream effector of NADPH oxidase with wortmannin and LY-294,002 also caused substantial inhibition of tamoxifen-induced elevation of [Ca(2+)](i). CONCLUSION: Tamoxifen increases [Ca(2+)](i) in human platelets by a non-genomic mechanism. Tamoxifen activates phospholipase Cγ as well as PI3-kinase and NADPH oxidase pathway to generate superoxide which causes the release of Ca(2+) from the endoplasmic reticulum, and promotes Ca(2+) influx into the platelets.


Subject(s)
Blood Platelets/drug effects , NADPH Oxidases/blood , Phosphatidylinositol 3-Kinases/blood , Selective Estrogen Receptor Modulators/pharmacology , Superoxides/blood , Tamoxifen/pharmacology , Blood Platelets/enzymology , Calcium/blood , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , Estrogen Antagonists/pharmacology , Female , Free Radical Scavengers/pharmacology , Humans , Male , NADPH Oxidases/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Phospholipase C gamma/blood , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/enzymology , Tamoxifen/analogs & derivatives , Time Factors , Up-Regulation
11.
J Clin Invest ; 121(11): 4462-76, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22005299

ABSTRACT

Diabetes mellitus is associated with platelet hyperactivity, which leads to increased morbidity and mortality from cardiovascular disease. This is coupled with enhanced levels of thromboxane (TX), an eicosanoid that facilitates platelet aggregation. Although intensely studied, the mechanism underlying the relationship among hyperglycemia, TX generation, and platelet hyperactivity remains unclear. We sought to identify key signaling components that connect high levels of glucose to TX generation and to examine their clinical relevance. In human platelets, aldose reductase synergistically modulated platelet response to both hyperglycemia and collagen exposure through a pathway involving ROS/PLCγ2/PKC/p38α MAPK. In clinical patients with platelet activation (deep vein thrombosis; saphenous vein graft occlusion after coronary bypass surgery), and particularly those with diabetes, urinary levels of a major enzymatic metabolite of TX (11-dehydro-TXB2 [TX-M]) were substantially increased. Elevated TX-M persisted in diabetic patients taking low-dose aspirin (acetylsalicylic acid, ASA), suggesting that such patients may have underlying endothelial damage, collagen exposure, and thrombovascular disease. Thus, our study has identified multiple potential signaling targets for designing combination chemotherapies that could inhibit the synergistic activation of platelets by hyperglycemia and collagen exposure.


Subject(s)
Aldehyde Reductase/blood , Blood Glucose/metabolism , Collagen/pharmacology , Platelet Activation/drug effects , Platelet Activation/physiology , Thromboxanes/blood , Adult , Aged , Aged, 80 and over , Aldehyde Reductase/antagonists & inhibitors , Aspirin/administration & dosage , Case-Control Studies , Diabetes Mellitus/blood , Enzyme Inhibitors/pharmacology , Female , Humans , In Vitro Techniques , Male , Middle Aged , Mitogen-Activated Protein Kinase 14/blood , Models, Biological , Oxidative Stress , Phospholipase C gamma/blood , Platelet Aggregation/drug effects , Platelet Aggregation/physiology , Platelet Aggregation Inhibitors/pharmacology , Protein Kinase C/blood , Reactive Oxygen Species/blood , Signal Transduction , Venous Thrombosis/blood
12.
J Biomed Sci ; 17: 45, 2010 Jun 04.
Article in English | MEDLINE | ID: mdl-20525309

ABSTRACT

BACKGROUND: 3-Hydroxy-3-methyl-glutaryl coenzyme A (HMG-CoA) reductase inhibitors (statins) have been widely used to reduce cardiovascular risk. These statins (i.e., simvastatin) may exert other effects besides from their cholesterol-lowering actions, including inhibition of platelet activation. Platelet activation is relevant to a variety of coronary heart diseases. Although the inhibitory effect of simvastatin in platelet activation has been studied; the detailed signal transductions by which simvastatin inhibit platelet activation has not yet been completely resolved. METHODS: The aim of this study was to systematically examine the detailed mechanisms of simvastatin in preventing platelet activation. Platelet aggregation, flow cytometric analysis, immunoblotting, and electron spin resonance studies were used to assess the antiplatelet activity of simvastatin. RESULTS: Simvastatin (20-50 microM) exhibited more-potent activity of inhibiting platelet aggregation stimulated by collagen than other agonists (i.e., thrombin). Simvastatin inhibited collagen-stimulated platelet activation accompanied by [Ca2+]i mobilization, thromboxane A2 (TxA2) formation, and phospholipase C (PLC)gamma2, protein kinase C (PKC), and mitogen-activated protein kinases (i.e., p38 MAPK, JNKs) phosphorylation in washed platelets. Simvastatin obviously increased both cyclic AMP and cyclic GMP levels. Simvastatin markedly increased NO release, vasodilator-stimulated phosphoprotein (VASP) phosphorylation, and endothelial nitric oxide synthase (eNOS) expression. SQ22536, an inhibitor of adenylate cyclase, markedly reversed the simvastatin-mediated inhibitory effects on platelet aggregation, PLCgamma2 and p38 MAPK phosphorylation, and simvastatin-mediated stimulatory effects on VASP and eNOS phosphorylation. CONCLUSION: The most important findings of this study demonstrate for the first time that inhibitory effect of simvastatin in platelet activation may involve activation of the cyclic AMP-eNOS/NO-cyclic GMP pathway, resulting in inhibition of the PLCgamma2-PKC-p38 MAPK-TxA2 cascade, and finally inhibition of platelet aggregation.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mitogen-Activated Protein Kinase Kinases/blood , Nucleotides, Cyclic/blood , Platelet Activation/drug effects , Platelet Activation/physiology , Simvastatin/pharmacology , Calcium Signaling/drug effects , Cell Adhesion Molecules/blood , Collagen/pharmacology , Cyclic AMP/blood , Cyclic GMP/blood , Humans , Hydroxyl Radical/blood , In Vitro Techniques , MAP Kinase Signaling System/drug effects , Microfilament Proteins/blood , Nitric Oxide/blood , Nitric Oxide Synthase Type III/blood , Phospholipase C gamma/blood , Phosphoproteins/blood , Platelet Aggregation Inhibitors/pharmacology , Protein Kinase C/blood , Signal Transduction/drug effects , Thromboxane A2/blood
13.
Arch Pharm Res ; 33(3): 387-94, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20361303

ABSTRACT

Paclitaxel is an anticancer drug used in the treatment of ovarian, breast, head and neck, lung, and prostate cancer. We investigated the antiplatelet activity of paclitaxel in vitro as well as a possible antiplatelet mechanism. Paclitaxel inhibited washed rabbit platelet aggregation induced by collagen in a concentration dependent manner, with an IC(50) of 59.7 +/- 3.5. However, it had little effect on platelet aggregation mediated by arachidonic acid, U46619, a thromboxane (TX) A(2) mimic, or thrombin, suggesting that paclitaxel may strongly inhibit collagen mediated signal transduction. In accordance with these findings, paclitaxel blocked collagen induced cytosolic calcium mobilization, arachidonic acid liberation, and serotonin secretion. In addition, it inhibited arachidonic acid mediated platelet aggregation by about 37% by interfering with TXA(2) synthase as measured by the formation of arachidonic acid mediated TXA(2) and prostaglandin D(2), as well as cyclooxygenase-1 and TXA(2) synthase activity assays. Taken together, these results point to a cellular mechanism for the antiplatelet activity of paclitaxel through the inhibition of TXA(2) synthase and cytosolic calcium mobilization. This may contribute to the beneficial effects of paclitaxel on the cardiovascular system.


Subject(s)
Blood Platelets/drug effects , Enzyme Inhibitors/pharmacology , Paclitaxel/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Thromboxane-A Synthase/antagonists & inhibitors , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Arachidonic Acid/metabolism , Blood Platelets/enzymology , Calcium/blood , Collagen/metabolism , Cyclooxygenase 1/blood , Dose-Response Relationship, Drug , Phospholipase C gamma/blood , Phosphorylation , Prostaglandin D2/blood , Rabbits , Serotonin/blood , Signal Transduction/drug effects , Thrombin/metabolism , Thromboxane A2/blood , Thromboxane-A Synthase/blood , Time Factors
14.
J Exp Med ; 205(8): 1775-88, 2008 Aug 04.
Article in English | MEDLINE | ID: mdl-18663126

ABSTRACT

Src homology 2 domain-containing leukocyte phosphoprotein of 76 kD (SLP76), an adaptor that plays a critical role in platelet activation in vitro, contains three N-terminal tyrosine residues that are essential for its function. We demonstrate that mice containing complementary tyrosine to phenylalanine mutations in Y145 (Y145F) and Y112 and Y128 (Y112/128F) differentially regulate integrin and collagen receptor signaling. We show that mutation of Y145 leads to severe impairment of glycoprotein VI (GPVI)-mediated responses while preserving outside-in integrin signaling. Platelets from Y112/128F mice, although having mild defects in GPVI signaling, exhibit defective actin reorganization after GPVI or alpha IIb beta 3 engagement. The in vivo consequences of these signaling defects correlate with the mild protection from thrombosis seen in Y112/128F mice and the near complete protection observed in Y145F mice. Using genetic complementation, we further demonstrate that all three phosphorylatable tyrosines are required within the same SLP76 molecule to support platelet activation by GPVI.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/physiology , Blood Platelets/physiology , Integrins/physiology , Phosphoproteins/chemistry , Phosphoproteins/physiology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Agammaglobulinaemia Tyrosine Kinase , Amino Acid Substitution , Animals , CD36 Antigens/physiology , Female , Genetic Complementation Test , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mutagenesis, Site-Directed , Phospholipase C gamma/blood , Phosphoproteins/deficiency , Phosphoproteins/genetics , Phosphorylation , Platelet Aggregation/genetics , Platelet Aggregation/physiology , Platelet Glycoprotein GPIIb-IIIa Complex/physiology , Protein-Tyrosine Kinases/blood , Signal Transduction , Thrombosis/blood , Thrombosis/etiology , Thrombosis/genetics , Tyrosine/chemistry
15.
Blood ; 110(2): 529-35, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17374738

ABSTRACT

Coronary artery thrombosis is often initiated by platelet activation on collagen-rich subendothelial layers in the disrupted atherosclerotic plaque. The activating platelet collagen receptor glycoprotein VI (GPVI) noncovalently associates with the Fc receptor gamma-chain (FcRgamma), which signals through its immunoreceptor-tyrosine-based activation motif (ITAM) via the adaptor LAT leading to the activation of phospholipase Cgamma2 (PLCgamma2). GPVI is a promising antithrombotic target as anti-GPVI antibodies induce the irreversible loss of the receptor from circulating platelets by yet undefined mechanisms in humans and mice and long-term antithrombotic protection in the latter. However, the treatment is associated with transient but severe thrombocytopenia and reduced platelet reactivity to thrombin questioning its clinical usefulness. Here we show that GPVI down-regulation occurs through 2 distinct pathways, namely ectodomain shedding or internalization/intracellular clearing, and that both processes are abrogated in mice carrying a point mutation in the FcRgamma-associated ITAM. In mice lacking LAT or PLCgamma2, GPVI shedding is abolished, but the receptor is irreversibly down-regulated through internalization/intracellular clearing. This route of GPVI loss is not associated with thrombocytopenia or altered thrombin responses. These results reveal the existence of 2 distinct signaling pathways downstream of the FcRgamma-ITAM and show that it is possible to uncouple GPVI down-regulation from undesired side effects with obvious therapeutic implications.


Subject(s)
Blood Platelets/physiology , Platelet Activation/physiology , Platelet Membrane Glycoproteins/genetics , Receptors, Collagen/genetics , Signal Transduction , Animals , Blood Platelets/cytology , Enzyme-Linked Immunosorbent Assay , Fibrinolysis , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Phospholipase C gamma/blood , Receptors, IgG/blood
16.
Thromb Res ; 120(2): 251-8, 2007.
Article in English | MEDLINE | ID: mdl-17055557

ABSTRACT

The integrin alpha(IIb)beta(3) plays a critical role in mediating clot retraction by platelets which is important in vivo in consolidating thrombus formation. Actin-myosin interaction is essential for clot retraction. In the present study, we demonstrate that the structurally distinct Src kinase inhibitors, PP2 and PD173952, significantly reduced the rate of clot retraction, but did not prevent it reaching completion. This effect was accompanied by abolition of alpha(IIb)beta(3)-dependent protein tyrosine phosphorylation, including PLCgamma2. A role for PLCgamma2 in mediating clot retraction was demonstrated using PLCgamma2-deficient murine platelets. Furthermore, platelet adhesion to fibrinogen leads to MLC phosphorylation through a pathway that is inhibited by PP2 and by the PLC inhibitor, U73122. These results demonstrate a partial role for Src kinase-dependent activation of PLCgamma2 and MLC phosphorylation in mediating clot retraction downstream of integrin alpha(IIb)beta(3).


Subject(s)
Clot Retraction/physiology , Phospholipase C gamma/blood , src-Family Kinases/blood , Animals , Blood Platelets/drug effects , Blood Platelets/physiology , Clot Retraction/drug effects , Humans , In Vitro Techniques , Mice , Models, Biological , Myosin Light Chains/blood , Phospholipase C gamma/deficiency , Phosphorylation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction , src-Family Kinases/antagonists & inhibitors
17.
Blood ; 108(8): 2596-603, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-16788103

ABSTRACT

Botrocetin (bt)-facilitated binding of von Willebrand factor (VWF) to the platelet membrane glycoprotein (GP) Ib-IX-V complex on platelets in suspension initiates a signaling cascade that causes alphaIIbbeta3 activation and platelet aggregation. Previous work has demonstrated that bt/VWF-mediated agglutination activates alphaIIbbeta3 and elicits ATP secretion in a thromboxane A2 (TxA2)-dependent manner. The signaling that results in TxA2 production was shown to be initiated by Lyn, enhanced by Src, and propagated through Syk, SLP-76, PI3K, PLCgamma2, and PKC. Here, we demonstrate that the signaling elicited by GPIb-mediated agglutination that results in TxA2 production is dependent on Bruton tyrosine kinase (Btk). The results demonstrate that Btk is downstream of Lyn, Syk, SLP-76, and PI3K; upstream of ERK1/2, PLCgamma2, and PKC; and greatly enhances Akt phosphorylation. The relationship(s), if any, between ERK1/2, PLCgamma2, and PKC were not elucidated. The requirement for Btk and TxA2 receptor function in GPIb-dependent arterial thrombosis was confirmed in vivo by characterizing blood flow in ferric chloride-treated mouse carotid arteries. These results demonstrate that the Btk family kinase, Tec, cannot provide the function(s) missing because of the absence of Btk and that Btk is essential for both bt/VWF-mediated agglutination-induced TxA2 production and GPIb-dependent stable arterial thrombus formation in vivo.


Subject(s)
Crotalid Venoms/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Protein-Tyrosine Kinases/metabolism , Thrombosis/blood , von Willebrand Factor/metabolism , Agammaglobulinaemia Tyrosine Kinase , Animals , Carotid Artery Thrombosis/blood , Carotid Artery Thrombosis/etiology , MAP Kinase Signaling System , Male , Mice , Mice, Inbred CBA , Mice, Knockout , Mice, Mutant Strains , Phospholipase C gamma/blood , Phosphorylation , Platelet Aggregation/physiology , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , Signal Transduction , Thrombosis/etiology , Thromboxane A2/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...