Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
J Thromb Haemost ; 16(6): 1211-1225, 2018 06.
Article in English | MEDLINE | ID: mdl-29575487

ABSTRACT

Essentials Phosphoinositide 3-kinase and MAPK pathways crosstalk via PDK1. PDK1 is required for adenosine diphosphate-induced platelet activation and thromboxane generation. PDK1 regulates RAF proto-oncogene Ser/Thr kinase (Raf1) activation in the MAPK pathway. Genetic ablation of PDK1 protects against platelet-dependent thrombosis in vivo. SUMMARY: Background Platelets are dynamic effector cells with functions that span hemostatic, thrombotic and inflammatory continua. Phosphoinositide-dependent protein kinase 1 (PDK1) regulates protease-activated receptor 4-induced platelet activation and thrombus formation through glycogen synthase kinase3ß. However, whether PDK1 also signals through the ADP receptor and its functional importance in vivo remain unknown. Objective To establish the mechanism of PDK1 in ADP-induced platelet activation and thrombosis. Methods We assessed the role of PDK1 on 2MeSADP-induced platelet activation by measuring aggregation, thromboxane generation and phosphorylation events in the presence of BX-795, which inhibits PDK1, or by using platelet-specific PDK1 knockout mice and performing western blot analysis. PDK1 function in thrombus formation was assessed with an in vivo pulmonary embolism model. Results PDK1 inhibition with BX-795 reduced 2-methylthio-ADP (2MeSADP)-induced aggregation of human and murine platelets by abolishing thromboxane generation. Similar results were observed in pdk1-/- mice. PDK1 was also necessary for the phosphorylation of mitogen-activated protein kinase kinase 1/2 (MEK1/2), extracellular signal-regulated kinase 1/2, and cytosolic phospholipase A2, indicating that PDK1 regulates an upstream kinase in the mitogen-activated protein kinase (MAPK) pathway. We next determined that this upstream kinase is Raf-1, a serine/threonine kinase that is necessary for the phosphorylation of MEK1/2, as pharmacological inhibition and genetic ablation of PDK1 were sufficient to prevent Raf1 phosphorylation. Furthermore, in vivo inhibition or genetic ablation of PDK1 protected mice from collagen/epinephrine-induced pulmonary embolism. Conclusion PDK1 governs thromboxane generation and thrombosis in platelets that are stimulated with 2MeSADP by regulating activation of the MAPK pathway.


Subject(s)
3-Phosphoinositide-Dependent Protein Kinases/metabolism , Blood Platelets/enzymology , Mitogen-Activated Protein Kinases/blood , Platelet Aggregation/drug effects , Proto-Oncogene Proteins c-raf/blood , Pulmonary Embolism/enzymology , Thrombosis/enzymology , Thromboxanes/blood , 3-Phosphoinositide-Dependent Protein Kinases/antagonists & inhibitors , 3-Phosphoinositide-Dependent Protein Kinases/blood , 3-Phosphoinositide-Dependent Protein Kinases/deficiency , 3-Phosphoinositide-Dependent Protein Kinases/genetics , Animals , Blood Platelets/drug effects , Disease Models, Animal , Humans , Mice, Knockout , Phosphorylation , Platelet Aggregation Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Mas , Pulmonary Embolism/blood , Pulmonary Embolism/genetics , Pulmonary Embolism/prevention & control , Pyrimidines/pharmacology , Signal Transduction , Thiophenes/pharmacology , Thrombosis/blood , Thrombosis/genetics , Thrombosis/prevention & control
2.
J Thromb Haemost ; 13(7): 1325-34, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25944668

ABSTRACT

BACKGROUND: Platelet secretion is critical to development of acute thrombotic occlusion. Platelet dense granules contain a variety of important hemostatically active substances. Nevertheless, biogenesis of platelet granules is poorly understood. OBJECTIVES: Serum- and glucocorticoid-inducible kinase 1 (SGK1) has been shown to be highly expressed in platelets and megakaryocytes, but its role in the regulation of platelet granule biogenesis and its impact on thrombosis has not been investigated so far. METHODS AND RESULTS: Electron microscopy analysis of the platelet ultrastructure revealed a significant reduction in the number and packing of dense granules in platelets lacking SGK1 (sgk1(-/-) ). In sgk1(-/-) platelets serotonin content was significantly reduced and activation-dependent secretion of ATP, serotonin and CD63 significantly impaired. In vivo adhesion after carotis ligation was significantly decreased in platelets lacking SGK1 and occlusive thrombus formation after FeCl3 -induced vascular injury was significantly diminished in sgk1(-/-) mice. Transcript levels and protein abundance of dense granule biogenesis regulating GTPase Rab27b were significantly reduced in sgk1(-/-) platelets without affecting Rab27b mRNA stability. In MEG-01 cells transfection with constitutively active (S422) (D) SGK1 but not with inactive (K127) (N) SGK1 significantly enhanced Rab27b mRNA levels. Sgk1(-/-) megakaryocytes show significantly reduced expression of Rab27b and serotonin/CD63 levels compared with sgk1(+/+) megakaryocytes. Proteome analysis identified nine further vesicular transport proteins regulated by SGK1, which may have an impact on impaired platelet granule biogenesis in sgk1(-/-) platelets independent of Rab27b. CONCLUSIONS: The present observations identify SGK1 as a novel powerful regulator of platelet dense granule biogenesis, platelet secretion and thrombus formation. SGK1 is at least partially effective because it regulates transcription of Rab27b in megakaryocytes.


Subject(s)
Blood Platelets/enzymology , Carotid Artery Injuries/enzymology , Cytoplasmic Granules/enzymology , Immediate-Early Proteins/blood , Platelet Activation , Protein Serine-Threonine Kinases/blood , Secretory Vesicles/enzymology , Thrombosis/enzymology , Adenosine Triphosphate/blood , Adenosine Triphosphate/metabolism , Animals , Blood Platelets/metabolism , Blood Platelets/ultrastructure , Carotid Artery Injuries/blood , Carotid Artery Injuries/genetics , Carotid Artery Injuries/pathology , Cells, Cultured , Cytoplasmic Granules/metabolism , Cytoplasmic Granules/ultrastructure , Disease Models, Animal , Female , Genotype , Immediate-Early Proteins/deficiency , Immediate-Early Proteins/genetics , Male , Megakaryocytes/enzymology , Megakaryocytes/metabolism , Mice, Knockout , Phenotype , Platelet Aggregation , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/metabolism , Secretory Vesicles/metabolism , Secretory Vesicles/ultrastructure , Serotonin/blood , Serotonin/metabolism , Signal Transduction , Tetraspanin 30/blood , Tetraspanin 30/metabolism , Thrombosis/blood , Thrombosis/genetics , Thrombosis/pathology , Time Factors , Transfection , Up-Regulation , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
3.
J Thromb Haemost ; 13(5): 719-28, 2015 May.
Article in English | MEDLINE | ID: mdl-25660395

ABSTRACT

BACKGROUND: Surface expression of stromal cell-derived factor-1 (SDF-1, CXCL12) on platelets is enhanced during ischemic events and plays an important role in peripheral homing of stem cells and myocardial repair mechanisms. SDF-1 effects are mediated through CXCR4 and CXCR7. Both CXCR4 and CXCR7 are surface expressed on human platelets and to a higher degree in patients with coronary artery disease (CAD) compared with healthy controls. In this study, we investigated the prognostic role of platelet CXCR4- and CXCR7 surface expression in patients with symptomatic CAD. METHODS AND RESULTS: In a cohort study, platelet surface expression of CXCR4 and CXCR7 was measured by using flow cytometry in 284 patients with symptomatic CAD at the time of percutaneous coronary intervention (PCI). The primary combined end point was defined as all-cause death and/or myocardial infarction (MI) during 12-month follow-up. Secondary end points were defined as the single events of all-cause death and MI. We found significant differences of CXCR4 values in patients who developed a combined end point compared with event-free patients (mean MFIAUTHOR: Please define MFI at first use. 3.17 vs. 3.44, 95% confidence interval [CI] 0.09-0.45) and in patients who subsequently died (mean MFI 3.10 vs. 3.42, 95% CI 0.09-0.56). In multivariate Cox regression analysis, lower platelet CXCR4 levels were independently and significantly associated with all-cause mortality (hazard ratio 0.24, 95% CI 0.07-0.87) and the primary combined end point of all-cause death and/or MI (hazard ratio 0.30, 95% CI 0.13-0.72). CONCLUSION: These findings highlight a potential prognostic value of platelet expression CXCR4 on clinical outcomes in patients with CAD.


Subject(s)
Blood Platelets/metabolism , Chemokine CXCL12/blood , Coronary Artery Disease/blood , Receptors, CXCR4/blood , Receptors, CXCR/blood , Aged , Coronary Artery Disease/pathology , Female , Humans , Male , Prognosis
4.
J Thromb Haemost ; 13(4): 619-30, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25556321

ABSTRACT

BACKGROUND: Platelet cytoskeletal reorganization is essential for platelet adhesion and thrombus formation in hemostasis and thrombosis. The Rho GTPases RhoA, Rac1 and Cdc42 are the main players in cytoskeletal dynamics of platelets and induce filopodia and lamellipodia formation and actin polymerization to strongly increase the platelet surface upon activation. Moreover, they are important for platelet secretion, integrin activation and arterial thrombus formation. OBJECTIVES: Rho GTPases are regulated by GTPase-activating proteins (GAPs) that stimulate their GTPase activity to terminate Rho signaling. The regulation of Rho GTPase activity in platelets is not well defined. Recently, we identified oligophrenin1 (OPHN1), a RhoGAP in platelets that exhibits strong GTPase-stimulating activity towards RhoA, Cdc42 and Rac1. RESULTS: In the present study we show for the first time, that deficiency of OPHN1 led to abnormal Rho activation and increased platelet cytoskeletal reorganization, including cell adhesion and lamellipodia formation on fibrinogen. Furthermore, platelets from ophn1(-/-) mice showed enhanced susceptibility to platelet activation with alterations in actin distribution and early release of granules. Platelet activation was enhanced following GPVI and PAR4 stimulation. This translated into elevated platelet thrombus formation and promoted arterial thrombosis under low shear conditions with altered hemostasis, as detected by tail bleeding time. CONCLUSIONS: The results of the present study identified OPHN1 as an important regulator of platelet cytoskeletal reorganization and demonstrate that abnormal regulation of Rho proteins leads to increased platelet adhesion and thrombus formation under low shear conditions in vitro and in vivo, suggesting a prothrombotic phenotype of mice critical for acute thrombotic occlusions.


Subject(s)
Blood Coagulation , Blood Platelets/enzymology , Cytoskeletal Proteins/deficiency , GTPase-Activating Proteins/deficiency , Nuclear Proteins/deficiency , Thrombosis/enzymology , rho GTP-Binding Proteins/blood , Animals , Cytoskeletal Proteins/genetics , Cytoskeleton/enzymology , Disease Models, Animal , Enzyme Activation , Female , GTPase-Activating Proteins/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/blood , Nuclear Proteins/genetics , Platelet Activation , Pseudopodia/enzymology , Signal Transduction , Thrombosis/blood , Thrombosis/genetics , Time Factors , cdc42 GTP-Binding Protein/blood , rac1 GTP-Binding Protein/blood , rhoA GTP-Binding Protein
5.
Acta Physiol (Oxf) ; 213(1): 181-90, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24947805

ABSTRACT

The serum- and glucocorticoid-inducible kinase 1 (SGK1) is expressed in megakaryocytes and circulating platelets. In megakaryocytes, SGK1 activates transcription factor nuclear factor kappa-B (NF-κB), which in turn stimulates expression of Orai1, a Ca(2+) channel protein accomplishing store-operated Ca(2+) enrty (SOCE). SGK1 enhances SOCE and several Ca(2+) -sensitive platelet functions, including degranulation, integrin αII b ß3 activation, phosphatidylserine exposure, aggregation and thrombus formation. As shown in other cell types, stimulators of SGK1 expression include ischaemia, oxidative stress, hyperglycaemia, advanced glycation end products (AGEs) and a variety of hormones such as glucocorticoids, mineralocorticoids, transforming growth factor beta (TGFß), interleukin 6 (IL-6), platelet-derived growth factor (PDGF), thrombin and endothelin. Thus, SGK1-sensitive Ca(2+) signalling may contribute to altered platelet function in several clinical conditions including inflammation, metabolic syndrome, diabetes mellitus and chronic renal failure. Nevertheless, further studies are needed defining the contribution of altered SGK1 expression and activity to physiology and pathophysiology of platelets.


Subject(s)
Blood Platelets/metabolism , Calcium Channels/metabolism , Glucocorticoids/metabolism , Hyperglycemia/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Calcium Signaling/physiology , Humans
6.
Thromb Haemost ; 110(5): 925-30, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23846758

ABSTRACT

Platelet secretion and aggregation as well as thrombus formation of blood platelets critically depend on increase of cytosolic Ca2+ concentration ([Ca2+]i) mainly resulting from intracellular Ca2+ release followed by store operated Ca2+ entry (SOCE) through Ca2+ release activated channels (CRAC). SOCE is in part accomplished by the pore forming unit Orai and its regulator stromal interaction molecule (STIM). Orai1 and STIM1 transcription is stimulated by NF-κB (nuclear factor kappa B). Serum- and glucocorticoid-inducible kinase 1 (SGK1) up-regulates NF-κB-activity in megakaryocytes and thus Orai1-expression and SOCE in platelets. SGK1 is thus a powerful regulator of platelet Ca2+-signalling and thrombus formation and presumably participates in the regulation of platelet activation by a variety of hormones as well as clinical conditions (e.g. type 2 diabetes or metabolic syndrome) associated with platelet hyperaggregability and increased risk of thromboocclusive events. SOCE in platelets is further regulated by scaffolding protein Homer and chaperone protein cyclophilin A (CyPA). Additional potential regulators of Orai1/STIM1 and thus SOCE in platelets include AMP activated kinase (AMPK), protein kinase A (PKA), reactive oxygen species, lipid rafts, pH and mitochondrial Ca2+ buffering. Future studies are required defining the significance of those mechanisms for platelet Orai1 abundance and function, for SOCE into platelets and for platelet function in cardiovascular diseases.


Subject(s)
Blood Platelets/metabolism , Calcium Channels/metabolism , Diabetes Mellitus, Type 2/immunology , Membrane Proteins/metabolism , Metabolic Syndrome/immunology , Neoplasm Proteins/metabolism , Animals , Calcium Signaling , Humans , Immediate-Early Proteins/metabolism , NF-kappa B/metabolism , ORAI1 Protein , Platelet Activation , Protein Serine-Threonine Kinases/metabolism , Reactive Oxygen Species/metabolism , Stromal Interaction Molecule 1
SELECTION OF CITATIONS
SEARCH DETAIL