Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Sci Rep ; 10(1): 914, 2020 01 22.
Article in English | MEDLINE | ID: mdl-31969609

ABSTRACT

The in vitro production of blood platelets for transfusion purposes is an important goal in the context of a sustained demand for controlled products free of infectious, immune and inflammatory risks. The aim of this study was to characterize human platelets derived from CD34+ progenitors and to evaluate their hemostatic properties. These cultured platelets exhibited a typical discoid morphology despite an enlarged size and expressed normal levels of the major surface glycoproteins. They aggregated in response to ADP and a thrombin receptor agonist peptide (TRAP). After infusion into NSG mice, cultured and native platelets circulated with a similar 24 h half-life. Notably, the level of circulating cultured platelets remained constant during the first two hours following infusion. During this period of time their size decreased to reach normal values, probably due to their remodeling in the pulmonary circulation, as evidenced by the presence of numerous twisted platelet elements in the lungs. Finally, cultured platelets were capable of limiting blood loss in a bleeding assay performed in thrombocytopenic mice. In conclusion, we show here that cultured platelets derived from human CD34+ cells display the properties required for use in transfusion, opening the way to clinical trials.


Subject(s)
Antigens, CD34 , Blood Platelets/physiology , Hemostasis , Platelet Aggregation , Platelet Transfusion , Stem Cells , Adenosine Diphosphate/pharmacology , Animals , Blood Platelets/metabolism , Cells, Cultured , Female , Glycoproteins/metabolism , In Vitro Techniques , Mice, Transgenic , Peptide Fragments/pharmacology , Platelet Aggregation/drug effects
2.
Sci Rep ; 8(1): 15808, 2018 10 25.
Article in English | MEDLINE | ID: mdl-30361531

ABSTRACT

Platelets are produced upon profound reorganization of mature megakaryocytes (MK) leading to proplatelet elongation and release into the blood stream, a process termed thrombopoiesis. This highly dynamic process requires microtubules (MT) reorganization by mechanisms that are still incompletely understood. Adenomatous polyposis coli (APC) is a microtubule plus-end tracking protein involved in the regulation of MT in a number of cell systems and its inactivation has been reported to alter hematopoiesis. The aim of our study was to investigate the role of APC in megakaryopoiesis and the final steps of platelet formation. Down-regulation of APC in cultured human MK by RNA interference increased endomitosis and the proportion of cells able to extend proplatelets (68.8% (shAPC1) and 52.5% (shAPC2) vs 28.1% in the control). Similarly an increased ploidy and amplification of the proplatelet network were observed in MK differentiated from Lin- cells of mice with APC-deficiency in the MK lineage. In accordance, these mice exhibited increased platelet counts when compared to wild type mice (1,323 ± 111 vs 919 ± 52 platelets/µL; n = 12 p 0.0033**). Their platelets had a normal size, ultrastructure and number of microtubules coils and their main functions were also preserved. Loss of APC resulted in lower levels of acetylated tubulin and decreased activation of the Wnt signaling pathway. Thus, APC appears as an important regulator of proplatelet formation and overall thrombopoiesis.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Blood Platelets/metabolism , Microtubules/metabolism , Acetylation , Adenomatous Polyposis Coli Protein/deficiency , Animals , Blood Platelets/ultrastructure , Cell Lineage , Cells, Cultured , Megakaryocytes/cytology , Megakaryocytes/metabolism , Megakaryocytes/ultrastructure , Mice, Inbred C57BL , Mice, Transgenic , Microtubules/ultrastructure , Platelet Count , Ploidies , Wnt Signaling Pathway
3.
J Thromb Haemost ; 14(7): 1470-9, 2016 07.
Article in English | MEDLINE | ID: mdl-27148783

ABSTRACT

UNLABELLED: Essentials A signaling role of glycoprotein (GP)Ibß is postulated but not formally demonstrated in platelets. Lentiviral-mediated rescue in knock-out mice can be used to evaluate GPIbß function in vivo. Transduction of the native subunit corrected the main defects associated with GPIb-IX deficiency Deletion of intracellular 159-170 segment increased thrombosis, 150-160 removal increased bleeding. SUMMARY: Background The platelet glycoprotein (GP)Ib-V-IX complex is required for normal hemostasis and megakaryopoiesis. A role in GPIb-dependent responses has been ascribed to the less well characterized GPIbß subunit using a specific antibody and GPIb-IX transfected cells. Objectives Our aim was to evaluate, in vivo, the role of the GPIbß in hemostasis and thrombosis. Methods GPIbß(null) Sca-1(+) progenitors transduced with viral particles harboring hGPIbß were transplanted into lethally irradiated GPIbß(-/-) recipient mice. Results hGPIbß transplanted into the bone marrow of GPIbß(null) mice rescued GPIb-IX expression in 97% of circulating platelets. These platelets efficiently bound von Willebrand factor (VWF) and extended filopodia on a VWF matrix, demonstrating the restoration of GPIb-dependent adhesive and signaling properties. These mice exhibited less severe macrothrombocytopenia and had normal tail bleeding times as compared with GPIbß(null) mice. This strategy was employed to manipulate and evaluate the role of the GPIbß intracellular domain. Removal of the membrane proximal segment (Δ(150-160) ) decreased GPIb-IX expression by 43%, confirming its involvement in receptor assembly and biosynthesis, and resulted in increased bleeding times and decreased thrombosis in a mechanical injury model in the aorta. On the other hand, deletion of the C-flanking 159-170 segment allowed normal GPIb-IX expression, VWF-dependent responses and bleeding times, but resulted in enhanced arterial thrombosis. Conclusion This pointed to a repressor role of GPIbß in thrombus formation in vivo that was not predicted in studies of heterologous cells. These results highlight the utility of this lentiviral strategy for the structure-function evaluation of GPIb-IX in platelets.


Subject(s)
Bernard-Soulier Syndrome/genetics , Gene Transfer Techniques , Platelet Glycoprotein GPIb-IX Complex/metabolism , Animals , Aorta/metabolism , Bleeding Time , Blood Platelets/metabolism , Bone Marrow Cells/cytology , Female , Gene Deletion , Gene Expression Regulation , Genetic Vectors , HEK293 Cells , Hemorrhage , Hemostasis , Humans , Lentivirus , Male , Megakaryocytes/cytology , Mice , Mice, Knockout , Microscopy, Electron, Scanning , Protein Domains , Thrombosis/metabolism , Transgenes , von Willebrand Factor/metabolism
4.
J Thromb Haemost ; 14(6): 1268-84, 2016 06.
Article in English | MEDLINE | ID: mdl-26991240

ABSTRACT

UNLABELLED: Essentials Information about the formation of the demarcation membrane system (DMS) is still lacking. We investigated the role of the cytoskeleton in DMS structuration in megakaryocytes. Cdc42/Pak-dependent F-actin remodeling regulates DMS organization for proper megakaryopoiesis. These data highlight the mandatory role of F-actin in platelet biogenesis. SUMMARY: Background Blood platelet biogenesis results from the maturation of megakaryocytes (MKs), which involves the development of a complex demarcation membrane system (DMS). Therefore, MK differentiation is an attractive model for studying membrane remodeling. Objectives We sought to investigate the mechanism of DMS structuration in relationship to the cytoskeleton. Results Using three-dimensional (3D) confocal imaging, we have identified consecutive stages of DMS organization that rely on F-actin dynamics to polarize membranes and nuclei territories. Interestingly, microtubules are not involved in this process. We found that the mechanism underlying F-actin-dependent DMS formation required the activation of the guanosine triphosphate hydrolase Cdc42 and its p21-activated kinase effectors (Pak1/2/3). Förster resonance energy transfer demonstrated that active Cdc42 was associated with endomembrane dynamics throughout terminal maturation. Inhibition of Cdc42 or Pak1/2/3 severely destructured the DMS and blocked proplatelet formation. Even though this process does not require containment within the hematopoietic niche, because DMS structuration was observed upon thrombopoietin-treatment in suspension, integrin outside-in signaling was required for Pak activation and probably resulted from secretion of extracellular matrix by MKs. Conclusions These data indicate a functional link, mandatory for MK differentiation, between actin dynamics, regulated by Cdc42/Pak1/2/3, and DMS maturation.


Subject(s)
Actins/metabolism , Megakaryocytes/metabolism , cdc42 GTP-Binding Protein/chemistry , cdc42 GTP-Binding Protein/metabolism , Animals , Blood Platelets/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Membrane/metabolism , Cytoskeleton/metabolism , Fluorescence Resonance Energy Transfer , Humans , Imaging, Three-Dimensional , Lentivirus , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Electron, Transmission , Signal Transduction , Thrombopoiesis , p21-Activated Kinases/metabolism
5.
J Thromb Haemost ; 13(5): 851-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25736522

ABSTRACT

BACKGROUND: At the end of maturation, megakaryocytes (MKs) form long cytoplasmic extensions called proplatelets (PPT). Enormous changes in cytoskeletal structures cause PPT to extend further, to re-localize organelles such as mitochondria and to fragment, leading to platelet release. Two non-muscle myosin IIs (NMIIs) are expressed in MKs; however, only NMII-A (MYH9), but not NMII-B (MYH10), is expressed in mature MKs and is implicated in PPT formation. OBJECTIVES: To provide in vivo evidence on the specific role of NMII-A and IIB in MK PPT formation. METHODS: We studied two transgenic mouse models in which non-muscle myosin heavy chain (NMHC) II-A was genetically replaced either by II-B or by a chimeric NMHCII that combined the head domain of II-A with the rod and tail domains of II-B. RESULTS AND CONCLUSIONS: This work demonstrates that the kinetic properties of NM-IIA, depending on the N-terminal domain, render NMII-A the better NMII candidate to control PPT formation. Furthermore, the carboxyl-terminal domain determines myosin II localization in the constriction region of PPT and is responsible for the specific role of NMII in platelet release.


Subject(s)
Blood Platelets/metabolism , Myosin Type II/metabolism , Animals , Mice , Mice, Inbred C57BL
6.
J Thromb Haemost ; 13(1): 115-25, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25393502

ABSTRACT

BACKGROUND: Transgenic mice expressing cre recombinase under the control of the platelet factor 4 (Pf4) promoter, in the context of a 100-kb bacterial artificial chromosome, have become a valuable tool with which to study genetic modifications in the platelet lineage. However, the specificity of cre expression has recently been questioned, and the time of its onset during megakaryopoiesis remains unknown. OBJECTIVES/METHODS: To characterize the expression of this transgene, we used double-fluorescent cre reporter mice. RESULTS: In the bone marrow, Pf4-cre-mediated recombination had occurred in all CD42-positive megakaryocytes as early as stage I of maturation, and in rare CD42-negative cells. In circulating blood, all platelets had recombined, along with only a minor fraction of CD45-positive cells. However, we found that all tissues contained recombined cells of monocyte/macrophage origin. When recombined, these cells might potentially modify the function of the tissues under particular conditions, especially inflammatory conditions, which further increase recombination in immune cells. Unexpectedly, a subset of epithelial cells from the distal colon showed signs of recombination resulting from endogenous Pf4-cre expression. This is probably the basis of the unexplained colon tumors developed by Apc(flox/flox) ;Pf4-cre mice, generated in a separate study on the role of Apc in platelet formation. CONCLUSION: Altogether, our results indicate early recombination with full penetrance in megakaryopoiesis, and confirm the value of Pf4-cre mice for the genetic engineering of megakaryocytes and platelets. However, care must be taken when investigating the role of platelets in processes outside hemostasis, especially when immune cells might be involved.


Subject(s)
Cell Lineage , Integrases/genetics , Megakaryocytes/metabolism , Platelet Factor 4/metabolism , Animals , Blood Platelets/metabolism , Cells, Cultured , Chromosomes, Artificial, Bacterial , Colon/cytology , Colon/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Gene Expression Regulation, Developmental , Genotype , Leukocytes/metabolism , Male , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Phenotype , Platelet Factor 4/genetics , Recombination, Genetic , Signal Transduction , Thrombopoiesis
7.
J Thromb Haemost ; 9(4): 779-89, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21261806

ABSTRACT

BACKGROUND: The FeCl(3)-induced vascular injury model is widely used to study thrombogenesis in vivo, but the processes leading to vascular injury and thrombosis are poorly defined. OBJECTIVES: The aim of our study was to better characterize the mechanisms of FeCl(3)-induced vascular injury and thrombus formation, in order to evaluate the pathophysiological relevance of this model. METHODS: FeCl(3) was applied at different concentrations (from 7.5% to 20%) and for different time periods (up to 5 min) to mouse carotid or mesenteric arteries. RESULTS: Under all the conditions tested, ultrastructural analysis revealed that FeCl(3) diffused through the vessel wall, resulting in endothelial cell denudation without exposure of the inner layers. Hence, only the basement membrane components were exposed to circulating blood cells and might have contributed to thrombus formation. Shortly after FeCl(3) application, numerous ferric ion-filled spherical bodies appeared on the endothelial cells. Interestingly, platelets could adhere to these spheres and form aggregates. Immunogold labeling revealed important amounts of tissue factor at their surface, suggesting that these spheres may play a role in thrombin generation. In vitro experiments indicated that FeCl(3) altered the ability of adhesive proteins, including collagen, fibrinogen and von Willebrand factor, to support platelet adhesion. Finally, real-time intravital microscopy showed no protection against thrombosis in GPVI-immunodepleted and ß(1)(-/-) mice, suggesting that GPVI and ß(1) integrins, known to be involved in initial platelet adhesion and activation, do not play a critical role in FeCl(3)-induced thrombus formation. CONCLUSION: This model should be used cautiously, in particular to study the earliest stage of thrombus formation.


Subject(s)
Carotid Arteries/pathology , Chlorides/toxicity , Ferric Compounds/toxicity , Thrombosis/drug therapy , Vascular Diseases/drug therapy , Animals , Anticoagulants/pharmacology , Carotid Arteries/ultrastructure , Disease Models, Animal , Mice , Mice, Inbred C57BL , Microscopy, Electron, Scanning , Platelet Aggregation Inhibitors/pharmacology
8.
J Thromb Haemost ; 8(10): 2243-51, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20695978

ABSTRACT

BACKGROUND: Inactivation of the mouse Myh9 gene (Myh9Δ) or its mutation in MYH9-related diseases leads to macrothrombocytopenia. Paradoxically, previous studies using in vitro differentiated megakaryocytes showed an increased capacity for proplatelet formation when myosin was absent or inhibited. METHODS: To explore the origin of the thrombocytopenia induced by myosin deficiency, we studied proplatelet formation using bone marrow explants of wild-type (WT) and Myh9Δ mouse where megakaryocytes have matured in their native environment. RESULTS AND DISCUSSION: A dramatic decrease in the number and complexity of proplatelets was observed in megakaryocytes from Myh9Δ mice, while inhibition of myosin activity by blebbistatin increased proplatelet formation from WT mature megakaryocytes. Moreover, Myh9Δ megakaryocytes had a smaller size than the WT cells. These data indicate that myosin deficiency acts negatively on proplatelet formation, probably by impairing in situ megakaryocyte maturation, while myosin activity is dispensable at the latest stage of proplatelet formation. In addition, ultrastructural examination of Myh9Δ bone marrow revealed an increased proportion of megakaryocytes exhibiting signs of non-apoptotic cell death as compared with the WT mice. CONCLUSION: These data indicate that thrombocytopenia in Myh9Δ mice results from defective development of megakaryocyte size, impaired proplatelet formation and increased cell death.


Subject(s)
Blood Platelets/cytology , Megakaryocytes/cytology , Mutation , Nonmuscle Myosin Type IIA/genetics , Thrombocytopenia/genetics , Animals , Bone Marrow/ultrastructure , Caspase 3/metabolism , Cell Death , Cell Lineage , Cell Survival , Female , Heterocyclic Compounds, 4 or More Rings/metabolism , In Situ Nick-End Labeling , Male , Mice , Microscopy, Electron, Transmission/methods , Myosin Heavy Chains , Thrombocytopenia/etiology
9.
J Thromb Haemost ; 8(10): 2262-72, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20633211

ABSTRACT

BACKGROUND: Immature dendritic cells (DCs) patrol the circulation, where they can uptake antigens. It has been reported that mature monocyte-derived DCs have the ability to interact with an activated platelet monolayer under high shear conditions (1500s(-1) ). OBJECTIVES: In this study, we investigated whether platelets can recruit immature myeloid DCs (CD1c(+) ) directly isolated from blood (BDCs) and if so, which receptors are involved. RESULTS: Using flow cytometry and electron microscopy, we showed that BDCs interact with activated but not resting platelets in suspension. Interaction was also observed after perfusing BDCs under low flow conditions (150 s(-1) ) through collagen-coated microcapillaries in which platelets had adhered and formed aggregates. No such interaction could be detected at higher shear rates. Whereas initial transient attachment required the exposure of P-selectin on activated platelets and PSGL-1 on BDCs, subsequent stationary adhesion was dependent on α(IIb) ß(3) and α(M) ß(2) integrins on platelets and BDCs, respectively. Moreover, during their transient interaction, BDCs preferentially removed platelets located at the outer margin of the thrombus in a P-selectin- and integrin-dependent manner. CONCLUSION: This study provides evidence for an interaction between activated platelets and immature myeloid DCs only under low shear conditions. This could be of importance for BDC maturation and antigen uptake during normal hemostasis or in the context of atherothrombosis at sites of reduced blood flow.


Subject(s)
Dendritic Cells/cytology , Myeloid Cells/cytology , Platelet Activation , Blood Platelets/cytology , Cell Adhesion , Flow Cytometry/methods , Humans , Macrophage-1 Antigen/metabolism , Membrane Glycoproteins/metabolism , Microscopy, Confocal/methods , Microscopy, Electron/methods , P-Selectin/metabolism , Platelet Adhesiveness , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Stress, Mechanical
10.
J Thromb Haemost ; 8(1): 173-84, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19874458

ABSTRACT

BACKGROUND: We previously described a model of laser-induced thrombosis in mesenteric arterioles with superficial and deep levels of injury producing a transient thrombus resolving within 2 min and a larger almost occlusive thrombus, respectively. Both types of lesion were sensitive to platelet GPIIb-IIIa and P2Y(12) inhibition, whereas only deep injuries were sensitive to thrombin blockade. OBJECTIVE: The aim of the present study was to use histologic methods and electron and intravital microscopy to characterize the lesions and thrombi and to extend our knowledge of the sensitivity of this model to genetic and pharmacologic inhibition. RESULTS: A superficial injury was found to detach the endothelial cells and expose a collagen III- and IV-rich subendothelium where platelets could adhere. Tissue factor and fibrin were not detected. Deeper penetration of the external elastic lamina occurred in deep injuries, with exposure of collagen I, III and IV. Here the thrombus was composed of platelets exhibiting a decreasing gradient of degranulation from the deepest lesion area to the surface. Fibrin was found close to the most activated platelets. Consistently, glycoprotein VI (GPVI)-collagen and GPIb-von Willebrand factor (VWF) interactions were found to be critical in superficial injuries. After deep lesion, thrombus formation was modestly reduced in GPVI-immunodepleted mice and still strongly inhibited in VWF(-/-) mice. Combined hirudin infusion and GPVI depletion further inhibited thrombosis after deep injury. CONCLUSIONS: This study confirms the feasibility of inducing arterial thrombosis with distinct levels of severity and establishes the central roles of collagen and VWF in thrombus formation after superficial injury. Collagen, VWF and thrombin all appear to contribute to thrombosis after deep arterial lesion.


Subject(s)
Blood Platelets/ultrastructure , Endothelium, Vascular/ultrastructure , Mesenteric Arteries/ultrastructure , Mesenteric Vascular Occlusion/pathology , Platelet Adhesiveness , Thrombosis/pathology , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Collagen Type I/metabolism , Collagen Type III/metabolism , Collagen Type IV/metabolism , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/injuries , Endothelium, Vascular/metabolism , Feasibility Studies , Fibrin/metabolism , Fibrinolytic Agents/administration & dosage , Hirudins/administration & dosage , Injections, Subcutaneous , Lasers, Gas , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/injuries , Mesenteric Arteries/metabolism , Mesenteric Vascular Occlusion/blood , Mesenteric Vascular Occlusion/etiology , Mesenteric Vascular Occlusion/prevention & control , Mice , Mice, Knockout , Platelet Adhesiveness/drug effects , Platelet Membrane Glycoproteins/deficiency , Platelet Membrane Glycoproteins/metabolism , Severity of Illness Index , Thrombosis/blood , Thrombosis/etiology , Thrombosis/prevention & control , Time Factors , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
11.
J Thromb Haemost ; 8(5): 1077-87, 2010 May.
Article in English | MEDLINE | ID: mdl-19694944

ABSTRACT

SUMMARY BACKGROUND: Circulating platelets are initially recruited at the site of vessel injury by von Willebrand factor (VWF) immobilized on collagen fibers. This process, mediated by the GPIb-V-IX complex, is accompanied by specific intracellular signaling leading to reorganization of the platelet actin cytoskeleton and extension of filopodia. OBJECTIVES/METHODS: To evaluate the GPIbalpha and GPIbbeta intracellular domains contribution to this signaling, we generated Chinese hamster ovary (CHO) cells expressing a GPIb-IX complex with mutant forms of the two subunits and we measured their ability to extend filopodia upon adhesion on a VWF matrix. RESULTS: Complete intracellular deletion or elimination of the filamin or the 14-3-3zeta binding sites in GPIbalpha did not prevent filopodia extension. In contrast, deletion of the juxtamembrane (Leu(150)-Arg(160)) or central (Ala(159)-Pro(170)) intracellular segment of GPIbbeta resulted in a 21% and 23% reduction in the number of cells extending filopodia, respectively. This occurred without decreasing adhesion efficiency or GPIb-IX association with filamin A or 14-3-3zeta. Alanine scanning mutagenesis of the Leu(150)-Pro(170) segment identified Arg(164), Leu(165), Leu(167), Thr(168) and Pro(170) as important residues for efficient filopodia formation. Surprisingly, mutation of the Ser(166) PKA phosphorylation site did not alter adhesion and shape change. A role for the GPIbbeta subunit was reinforced by the decreased capacity to extend filopodia upon adhesion on VWF of platelets from knock-in mice expressing a GPIbbeta intracellular deletion mutant. CONCLUSIONS: Altogether, our results strongly support participation of GPIbbeta and its intracellular region in GPIb-dependent platelet activation and shape change triggered by a VWF matrix.


Subject(s)
Platelet Membrane Glycoproteins/metabolism , Serine/metabolism , von Willebrand Factor/metabolism , Animals , CHO Cells , Cell Adhesion , Cricetinae , Cricetulus , Humans , Mice , Mutagenesis , Phosphorylation , Platelet Membrane Glycoproteins/chemistry
12.
Arterioscler Thromb Vasc Biol ; 27(1): 241-7, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17095718

ABSTRACT

OBJECTIVE: The platelet glycoprotein (GP)Ib-V-IX complex is a receptor required for normal hemostasis deficient in the Bernard-Soulier bleeding disorder. To evaluate the consequences of GPIb-V-IX deficiency in thrombosis we generated mouse models of the disease by targeting the GPIb beta subunit. METHODS AND RESULTS: Complete deletion (GPIb beta-/-) or an intracellular truncation (GPIb beta deltaIC-/-) reproduced typical and variant forms of Bernard-Soulier, with absent and partial (20%) expression of the complex on the platelet surface. Both strains exhibited thrombocytopenia and enlarged platelets with abnormal microtubular structures but normal granule composition. They exhibited prolonged tail bleeding times, which were less pronounced in GPIb beta deltaIC-/-. Decreased thrombus formation was observed after blood perfusion over a collagen coated surface at high shear. Resistance to vascular occlusion and an abnormal thrombus composition were observed in a model of FeCl3-induced lesion of carotid arteries. In a model of laser-induced lesion of mesenteric arterioles, thrombosis was strongly reduced in GPIb beta-/- mice, while a more modest effect was observed in GPIb beta deltaIC-/- animals. Finally, the two strains were protected against death in a model of systemic thromboembolism. CONCLUSIONS: This study provides in vivo evidence of a decreased thrombotic tendency linked to defective platelet GPIb-V-IX in mouse models of Bernard-Soulier syndrome.


Subject(s)
Bernard-Soulier Syndrome/complications , Platelet Glycoprotein GPIb-IX Complex/metabolism , Thrombosis/etiology , Thrombosis/prevention & control , Animals , Bernard-Soulier Syndrome/metabolism , Bernard-Soulier Syndrome/physiopathology , Blood Platelets/pathology , Blood Platelets/physiology , Collagen , Disease Models, Animal , Gene Expression Regulation , Hemostasis/genetics , Hemostasis/physiology , Mice , Mice, Knockout , Platelet Count , Platelet Glycoprotein GPIb-IX Complex/genetics , Thrombocytopenia/pathology , Thrombocytopenia/physiopathology , Thrombosis/metabolism , Thrombosis/physiopathology
13.
J Thromb Haemost ; 4(12): 2645-55, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17100656

ABSTRACT

BACKGROUND: Interaction between the platelet glycoprotein (GP)Ib-V-IX complex and von Willebrand factor (VWF) is critical for initiating platelet-vessel wall contacts, particularly under high shear conditions. This interaction also plays an important role in initiating platelet activation through the generation of intracellular signals resulting in platelet shape change and integrin alpha(IIb)beta3 activation. OBJECTIVE: A cell-penetrating peptide strategy was used to study the role of the intracellular domain of the GPIbalpha subunit in VWF/GPIb-V-IX-dependent adhesion and activation. METHODS: Peptides of 11-13 amino acids, covering the 557-610 region, were coupled to a nine-arginine permeating tag (R9) and the effects of their cell entry on VWF-dependent responses were analyzed. RESULTS: The R9alpha557 peptide corresponding to the 557-569 segment reduced platelet agglutination in response to VWF, while the other peptides had no effect. The decreased platelet agglutination appeared to be an indirect consequence of adenosine diphosphate release as a normal response was restored by apyrase or a P2Y1 receptor antagonist. A more direct effect of R9alpha557 on GPIb VWF-dependent functions was observed in adhesion studies on a VWF matrix, where it decreased platelet adhesion and profoundly inhibited filopodia formation. In addition, cell adhesion was reduced and shape change absent when Chinese hamster ovary cells expressing the GPIb-IX complex were incubated with R9alpha557. CONCLUSION: This study performed in intact platelets suggests a functional role of the 557-569 domain of GPIbalpha in controlling VWF-dependent adhesion and signaling.


Subject(s)
Blood Platelets/metabolism , Membrane Proteins/metabolism , Platelet Adhesiveness , Platelet Aggregation , Platelet Glycoprotein GPIb-IX Complex/metabolism , Signal Transduction , Adenosine Diphosphate/metabolism , Animals , Binding Sites , Blood Platelets/cytology , CHO Cells , Cell Adhesion , Cell Shape , Cricetinae , Cricetulus , Flow Cytometry , Humans , In Vitro Techniques , Membrane Glycoproteins , Membrane Proteins/chemistry , Membrane Proteins/genetics , Peptides/chemical synthesis , Peptides/metabolism , Platelet Glycoprotein GPIb-IX Complex/chemistry , Platelet Glycoprotein GPIb-IX Complex/genetics , Protein Binding , Protein Structure, Tertiary , Stress, Mechanical , Transfection , von Willebrand Factor/metabolism
14.
J Thromb Haemost ; 4(1): 217-28, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16409472

ABSTRACT

The molecular defect of a new Bernard-Soulier patient, originating from Morocco and presenting thrombocytopenia with large platelets and an absence of ristocetin-induced platelet agglutination, has been identified and reproduced in transfected heterologous cells. Gene sequencing revealed insertion of a guanine in the domain coding for the transmembrane region of the glycoprotein (GP) Ib beta subunit. This mutation causes a translational frame shift, which creates putative novel transmembrane and cytoplasmic 37 and 125 amino acids domains, respectively. A 34 kDa immunoreactive GPIb beta band, instead of the normal 26 kDa subunit, was detected by Western blotting in lysates from the patient's platelets and from transfected cells and in immunoprecipitates of metabolically labeled cells. The abnormal subunit did not associate with GPIb alpha and was mainly intracellular, although a significant fraction could reach the cell surface. Cells expressing the mutant GPIb-IX complex adhered to a von Willebrand factor matrix but were unable to change shape, unlike cells expressing the wild-type receptor. These results strongly suggest a novel role of the GPIb beta subunit and its transmembrane-intracellular region in GPIb-VWF-dependent signaling, in addition to a role in correct assembly and cell surface targeting of the GPIb-V-IX complex.


Subject(s)
Bernard-Soulier Syndrome/genetics , Platelet Glycoprotein GPIb-IX Complex/genetics , Signal Transduction , Amino Acid Sequence , Animals , Bernard-Soulier Syndrome/blood , Bernard-Soulier Syndrome/complications , Blood Platelets/pathology , CHO Cells , Cell Membrane , Cell Shape , Child, Preschool , Cricetinae , Cytoplasm , Female , Frameshift Mutation , Humans , Peptide Fragments , Signal Transduction/genetics , Thrombocytopenia/etiology , Transfection , von Willebrand Factor/metabolism
16.
J Thromb Haemost ; 2(6): 969-77, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15140133

ABSTRACT

ADP and TxA2 are secondary agonists which play an important role as cofactors when platelets are activated by agonists such as collagen or thrombin. The aim of the present study was to characterize the role of the ADP receptor P2Y(1) in collagen-induced activation of washed platelets. Inhibition of P2Y(1) alone with the selective antagonist MRS2179 prolonged the lag phase preceding aggregation in response to low or high concentrations of fibrillar collagen, without affecting the maximum amplitude of aggregation or secretion. A combination of MRS2179 and aspirin resulted in complete inhibition of platelet shape change at low and high collagen concentrations, together with a profound decrease in aggregation and secretion. Scanning electron microscopy showed that these platelets had conserved the discoid morphology typical of the resting state. A lack of shape change was also observed in aspirin-treated P2Y(1)- and G(alphaq)-deficient mouse platelets and in delta-storage pool-deficient platelets from Fawn Hooded rats. In contrast, when the second ADP receptor P2Y(12) was inhibited with AR-C69931MX, aspirin-treated platelets were still able to change shape and displayed only a moderate decrease in aggregation and secretion. In conclusion, this study provides evidence that collagen requires not only the TxA2 receptor Tpalpha, but also P2Y(1), to induce platelet shape change.


Subject(s)
Adenosine Diphosphate/analogs & derivatives , Blood Platelets/cytology , Collagen/pharmacology , Receptors, Purinergic P2/physiology , Adenosine Diphosphate/metabolism , Adenosine Diphosphate/pharmacology , Animals , Aspirin/pharmacology , Blood Platelets/metabolism , Blood Platelets/physiology , Cell Shape/drug effects , Drug Synergism , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Kinetics , Mice , Mice, Knockout , Microscopy, Electron, Scanning , Platelet Activation , Purinergic P2 Receptor Antagonists , Rats , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2Y1 , Signal Transduction , Thromboxane A2/antagonists & inhibitors , Thromboxane A2/biosynthesis
17.
J Thromb Haemost ; 1(1): 155-63, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12871553

ABSTRACT

In order to investigate the role of the platelet P2Y1 receptor in several aspects of platelet activation and thrombosis, transgenic (TG) mice overexpressing this receptor specifically in the megakaryocytic/platelet lineage were generated using the promoter of the tissue-specific platelet factor 4 gene. Studies of the saturation binding of [33P]2MeSADP in the presence or absence of the selective P2Y1 antagonist MRS2179 indicated that wild-type (WT) mouse platelets bore 150 +/- 31 P2Y1 receptors and TG platelets 276 +/- 34, representing an 84% increase in P2Y1 receptor density. This led to a well defined phenotype of platelet hyper-reactivity in vitro, as shown by increased aggregations in response to adenosine 5'-diphosphate (ADP) and low concentration of collagen in TG as compared with WT platelets. Moreover, overexpression of the P2Y1 receptor enabled ADP to induce granule secretion, unlike in WT platelets, which suggests that the level of P2Y1 expression is critical for this event. Our results further suggest that the weak responses of normal platelets to ADP are due to a limited number of P2Y1 receptors rather than to activation of a specific transduction pathway. TG mice displayed a shortened bleeding time and an increased sensitivity to in vivo platelet aggregation induced by infusion of a mixture of collagen and epinephrine. Overall, these findings emphasize the importance of the P2Y1 receptor in hemostasis and thrombosis and suggest that variable expression levels of this receptor on platelets might play a role in thrombotic states in human, which remains to be assessed.


Subject(s)
Blood Platelets/metabolism , Cell Lineage/genetics , Platelet Activation/physiology , Receptors, Purinergic P2/biosynthesis , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/metabolism , Adenosine Diphosphate/pharmacology , Animals , Bleeding Time , Calcium/analysis , Calcium/chemistry , Calcium/metabolism , Collagen/pharmacology , Cyclic AMP/analysis , Epinephrine/pharmacology , Gene Expression , Male , Mice , Mice, Transgenic , Plasmids/genetics , Platelet Aggregation/physiology , Platelet Factor 4/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Purinergic P2 Receptor Antagonists , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2Y1
18.
FEBS Lett ; 542(1-3): 53-9, 2003 May 08.
Article in English | MEDLINE | ID: mdl-12729897

ABSTRACT

The role of the phospholipase C (PLC)gamma 2 isotype in platelet activation was evaluated by studying PLC gamma 2 -/- mice. These mice have a prolonged bleeding time but their platelets respond normally to non-collagenous agonists. PLC gamma 2-null platelets show residual aggregation response to collagen fibres (6% versus 74% for wild-type) with minimal granule secretion and no shape change. A delayed shape change is observed at later aggregation times. Specific activation by glycoprotein (GP)VI agonists (convulxin, collagen-related peptide and GPVI crosslinking) is, however, abolished. Antibodies against integrin alpha(2)beta(1) and GPVI each inhibit the residual collagen response, implying a role of alpha(2)beta(1) in platelet activation and a functional association with GPVI. These responses are also prevented by blocking integrin alpha(IIb)beta(3) and phosphoinositide 3-kinase, whereas aspirin treatment and ADP receptor blockade only inhibit shape change. These results provide evidence for a PLC gamma 2-independent collagen activation pathway requiring cooperation between GPVI and alpha(2)beta(1) leading to alpha(IIb)beta(3)-dependent aggregation and shape change by released ADP and thromboxane A(2).


Subject(s)
Collagen/pharmacology , Integrin alpha2beta1/physiology , Platelet Aggregation , Platelet Membrane Glycoproteins/physiology , Type C Phospholipases/physiology , Animals , Bleeding Time , Blood Platelets/cytology , Blood Platelets/enzymology , Blood Platelets/metabolism , Blood Platelets/physiology , Integrin alpha2beta1/metabolism , Ligands , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Phospholipase C gamma , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Membrane Glycoproteins/metabolism , Secretory Vesicles/metabolism , Type C Phospholipases/genetics
19.
FEBS Lett ; 505(2): 281-90, 2001 Sep 14.
Article in English | MEDLINE | ID: mdl-11566191

ABSTRACT

High concentrations of adenosine-5'-diphosphate ADP are able to induce partial aggregation without shape change of P2Y(1) receptor-deficient mouse platelets through activation of the P2Y(12) receptor. In the present work we studied the transduction pathways selectively involved in this phenomenon. Flow cytometric analyses using R-phycoerythrin-conjugated JON/A antibody (JON/A-PE), an antibody which recognizes activated mouse alpha(IIb)beta(3) integrin, revealed a low level activation of alpha(IIb)beta(3) in P2Y(1) receptor-deficient platelets in response to 100 microM ADP or 1 microM 2MeS-ADP. Adrenaline induced no such activation but strongly potentiated the effect of ADP in a dose-dependent manner. Global phosphorylation of (32)P-labeled platelets showed that P2Y(12)-mediated aggregation was not accompanied by an increase in the phosphorylation of myosin light chain (P(20)) or pleckstrin (P(47)) and was not affected by the protein kinase C (PKC) inhibitor staurosporine. On the other hand, two unrelated phosphoinositide 3-kinase inhibitors, wortmannin and LY294002, inhibited this aggregation. Our results indicate that (i) the P2Y(12) receptor is able to trigger a P2Y(1) receptor-independent inside-out signal leading to alpha(IIb)beta(3) integrin activation and platelet aggregation, (ii) ADP and adrenaline use different signaling pathways which synergize to activate the alpha(IIb)beta(3) integrin, and (iii) the transduction pathway triggered by the P2Y(12) receptor is independent of PKC but dependent on phosphoinositide 3-kinase.


Subject(s)
Membrane Proteins , Phosphatidylinositol 3-Kinases/metabolism , Platelet Aggregation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Receptors, Purinergic P2/physiology , Androstadienes/pharmacology , Animals , Blood Proteins/metabolism , Chromones/pharmacology , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , Epinephrine/pharmacology , Fibrinogen/metabolism , Flow Cytometry , Kinetics , Mice , Microscopy, Electron, Scanning , Morpholines/pharmacology , Myosin Light Chains/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2Y12 , Signal Transduction , Staurosporine/pharmacology , Time Factors , Wortmannin
20.
Blood ; 97(12): 3829-35, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11389023

ABSTRACT

Collagen-induced platelet aggregation is a complex process and involves synergistic action of integrins, immunoglobulin (Ig)-like receptors, G-protein-coupled receptors and their ligands, most importantly collagen itself, thromboxane A(2) (TXA(2)), and adenosine diphosphate (ADP). The precise role of each of these receptor systems in the overall processes of activation and aggregation, however, is still poorly defined. Among the collagen receptors expressed on platelets, glycoprotein (GP) VI has been identified to play a crucial role in collagen-induced activation. GPVI is associated with the FcRgamma chain, which serves as the signal transducing unit of the receptor complex. It is well known that clustering of GPVI by highly specific agonists results in platelet activation and irreversible aggregation, but it is unclear whether collagen has the same effect on the receptor. This study shows that platelets from Galphaq-deficient mice, despite their severely impaired response to collagen, normally aggregate on clustering of GPVI, suggesting this not to be the principal mechanism by which collagen activates platelets. On the other hand, dimerization of GPVI by a monoclonal antibody (JAQ1), which by itself did not induce aggregation, provided a sufficient stimulus to potentiate platelet responses to Gi-coupled, but not Gq-coupled, agonists. The combination of JAQ1 and adrenaline or ADP, but not serotonin, resulted in alpha(IIb)beta(3)-dependent aggregation that occurred without intracellular calcium mobilization and shape change in the absence of Galphaq or the P2Y(1) receptor. Together, these results provide evidence for a cross-talk between (dimerized) GPVI and Gi-coupled receptors during collagen-induced platelet aggregation. (Blood. 2001;97:3829-3835)


Subject(s)
Heterotrimeric GTP-Binding Proteins/metabolism , Membrane Proteins , Platelet Membrane Glycoproteins/metabolism , Receptor Cross-Talk/physiology , Animals , Antibodies, Monoclonal/pharmacology , Collagen/pharmacology , Drug Synergism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , GTP-Binding Protein alpha Subunits, Gq-G11 , Heterotrimeric GTP-Binding Proteins/deficiency , Heterotrimeric GTP-Binding Proteins/genetics , Mice , Mice, Knockout , Platelet Aggregation/drug effects , Platelet Aggregation/physiology , Platelet Membrane Glycoproteins/immunology , Platelet Membrane Glycoproteins/physiology , Receptor Aggregation/drug effects , Receptor Aggregation/physiology , Receptors, Purinergic P2/drug effects , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2Y12 , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...