Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
Cell Signal ; 26(9): 1975-84, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24703939

ABSTRACT

Cytoskeletal reorganization is crucial for platelet adhesion and thrombus formation to avoid excessive bleeding. Major regulators of cytoskeletal dynamics are small GTPases of the Rho family. Rho GTPases become activated by G-protein coupled receptor activation, downstream of immunoreceptor tyrosine-based activation motif (ITAM)-coupled receptors and by outside-in signaling of integrins. They act as molecular switches and cycle between active and inactive states. GTPase activating proteins (GAPs) stimulate the hydrolysis of GTP to GDP to terminate Rho signaling. Nadrin is a RhoGAP that was recently identified in platelets. Five Nadrin isoforms are known consisting of a unique GAP and an N-terminal BAR domain responsible for the selective regulation of RhoA, Cdc42 and Rac1. Besides BAR domain mediated regulation of Nadrin GAP activity nothing is known about the regulation of Nadrin and the impact on cytoskeletal reorganization. Here we show that Nadrin becomes tyrosine phosphorylated upon platelet activation. We found Src family proteins (Src, Lyn, Fyn) to be responsible to control Nadrin GAP activity by phosphorylation. Interestingly, phosphorylation of Nadrin leads to tightly regulated Rho activation that was found to be Nadrin isoform- and (Rho) target-specific. Src-phosphorylation of Nadrin5 mediated inactivation of Cdc42 while RhoA and Rac1 became activated upon Src-mediated phosphorylation of Nadrin2. Our results suggest a critical role for spatial and temporal regulation of Nadrin and thus for the control of Rho GTPases in platelets.


Subject(s)
Blood Platelets/metabolism , GTPase-Activating Proteins/metabolism , Tyrosine/metabolism , Animals , CHO Cells , Cell Adhesion , Cricetinae , Cricetulus , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Phosphorylation , Platelet Activation/drug effects , Protein Isoforms/metabolism , Thrombin/pharmacology , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , src-Family Kinases/metabolism
3.
Phys Rev Lett ; 111(17): 172501, 2013 Oct 25.
Article in English | MEDLINE | ID: mdl-24206482

ABSTRACT

The nucleus (154)Gd is located in a region of the nuclear chart where rapid changes of nuclear deformation occur as a function of particle number. It was investigated using a combination of γ-ray scattering experiments and a γγ-coincidence study following electron capture decay of (154)Tb(m). A novel decay channel from the scissors mode to the first excited 0(+) state was observed. Its transition strength was determined to B(M1;1(sc)(+)→0(2)(+))=0.031(4)µ(N)(2). The properties of the scissors mode of (154)Gd imply a much larger matrix element than previously thought for the neutrinoless double-ß decay to the 0(2)(+) state in such a shape-transitional region. Theory indicates an even larger effect for (150)Nd.

4.
Cell Signal ; 25(1): 236-46, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22975681

ABSTRACT

Cytoskeletal reorganization of activated platelets plays a crucial role in hemostasis and thrombosis and implies activation of Rho GTPases. Rho GTPases are important regulators of cytoskeletal dynamics and function as molecular switches that cycle between an inactive and an active state. They are regulated by GTPase activating proteins (GAPs) that stimulate GTP hydrolysis to terminate Rho signaling. The regulation of Rho GTPases in platelets is not explored. A detailed characterization of Rho regulation is necessary to understand activation and inactivation of Rho GTPases critical for platelet activation and aggregation. Nadrin is a RhoGAP regulating cytoplasmic protein explored in the central nervous system. Five Nadrin isoforms are known that share a unique GAP domain, a serine/threonine/proline-rich domain, a SH3-binding motif and an N-terminal BAR domain but differ in their C-terminus. Here we identified Nadrin in platelets where it co-localizes to actin-rich regions and Rho GTPases. Different Nadrin isoforms selectively regulate Rho GTPases (RhoA, Cdc42 and Rac1) and cytoskeletal reorganization suggesting that - beside the GAP domain - the C-terminus of Nadrin determines Rho specificity and influences cell physiology. Furthermore, Nadrin controls RhoA-mediated stress fibre and focal adhesion formation. Spreading experiments on fibrinogen revealed strongly reduced cell adhesion upon Nadrin overexpression. Unexpectedly, the Nadrin BAR domain controls Nadrin-GAP activity and acts as a guidance domain to direct this GAP to its substrate at the plasma membrane. Our results suggest a critical role for Nadrin in the regulation of RhoA, Cdc42 and Rac1 in platelets and thus for platelet adhesion and aggregation.


Subject(s)
Blood Platelets/metabolism , GTPase-Activating Proteins/metabolism , Actins/metabolism , Animals , CHO Cells , Cell Adhesion , Cricetinae , Cricetulus , Cytoskeleton , GTPase-Activating Proteins/chemistry , Mice , Mice, Inbred C57BL , Platelet Activation , Platelet Aggregation , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Structure, Tertiary , Transfection , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
5.
J Thromb Haemost ; 10(6): 1152-64, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22448969

ABSTRACT

BACKGROUND: Bone-marrow-derived progenitor cells are important in myocardial repair mechanisms following prolonged ischemia. Cell-based therapy of diseased myocardium is limited by a low level of tissue engraftment. OBJECTIVES: The aim of this study was the development of the bifunctional protein αCD133-glycoprotein (GP)VI as an effective treatment for supporting vascular and myocardial repair mechanisms. RESULTS: We have generated and characterized a bifunctional molecule (αCD133-GPVI) that binds both to the subendothelium of the injured microvasculature and to CD133(+) progenitor cells with high affinity. αCD133-GPVI enhances progenitor cell adhesion to extracellular matrix proteins and differentiation into mature endothelial cells. In vivo studies showed that αCD133-GPVI favors adhesion of circulating progenitor cells to the injured vessel wall (intravital microscopy). Also, treatment of mice undergoing experimental myocardial infarction with αCD133-GPVI-labeled progenitor cells reduces infarction size and preserves myocardial function. CONCLUSIONS: The bifunctional trapping protein αCD133-GPVI represents a novel and promising therapeutic option for limiting heart failure of the ischemic myocardium.


Subject(s)
Antigens, CD/immunology , Endothelial Cells/transplantation , Genetic Therapy , Glycoproteins/immunology , Myocardial Infarction/therapy , Myocardium/pathology , Peptides/immunology , Platelet Membrane Glycoproteins/biosynthesis , Regeneration , Single-Chain Antibodies/biosynthesis , Stem Cell Transplantation , AC133 Antigen , Animals , Binding Sites , Cell Adhesion , Cell Differentiation , Disease Models, Animal , Endothelial Cells/immunology , Endothelial Cells/metabolism , Extracellular Matrix Proteins/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Myocardial Infarction/genetics , Myocardial Infarction/immunology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/immunology , Myocardium/metabolism , Platelet Membrane Glycoproteins/genetics , Recombinant Proteins/biosynthesis , Single-Chain Antibodies/genetics , Time Factors , Transfection , Ventricular Function, Left
6.
J Thromb Haemost ; 8(6): 1353-63, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20230420

ABSTRACT

BACKGROUND: Agonist-induced platelet activation involves different signaling pathways leading to the activation of phospholipase C (PLC) beta or PLCgamma2. Activated PLC produces inositol 1,4,5-trisphosphate and diacylglycerol, which trigger Ca(2+) mobilization and the activation of protein kinase C, respectively. PLCbeta is activated downstream of Gq-coupled receptors for soluble agonists with only short interaction times in flowing blood. In contrast, PLCgamma2 becomes activated downstream of receptors that interact with immobilized ligands such as the collagen receptor glycoprotein (GP) VI or activated integrins. OBJECTIVE AND METHODS: We speculated that PLCgamma2 activity might be optimized for sustained but submaximal signaling to control relatively slow platelet responses. To test this hypothesis, we analyzed platelets from mice heterozygous for a gain-of-function mutation in the Plcg2 gene (Plcg2(Ali5/+)). RESULTS: Plcg2(Ali5/+) platelets showed enhanced Ca(2+) mobilization, integrin activation, granule secretion and phosphatidylserine exposure upon GPVI or C-type lectin-like receptor-2 stimulation. Furthermore, integrin alpha(IIb)beta(3) outside-in signaling was markedly enhanced in the mutant platelets, as shown by accelerated spreading on different matrices and faster clot retraction. These defects translated into virtually unlimited thrombus formation on collagen under flow in vitro and a prothrombotic phenotype in vivo. CONCLUSIONS: These results demonstrate that the enzymatic activity of PLCgamma2 is tightly regulated to ensure efficient but limited platelet activation at sites of vascular injury.


Subject(s)
Blood Platelets/physiology , Mutation , Phospholipase C gamma/metabolism , Thrombosis/genetics , Animals , Blood Platelets/enzymology , Flow Cytometry , Heterozygote , Mice , Phenotype , Phospholipase C gamma/genetics , Platelet Aggregation
7.
J Thromb Haemost ; 7 Suppl 1: 206-9, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19630801

ABSTRACT

Heterodimeric receptors of the beta1 and beta3 integrin families mediate platelet adhesion and aggregation in hemostasis and thrombosis. In resting platelets, integrins are expressed in a low-affinity state but they shift to a high-affinity state and efficiently bind their ligands in response to cellular activation. This review summarizes recent advances in understanding the functional regulation and (patho-) physiological significance of individual platelet integrins with a special focus on studies in genetically modified mice. It is now recognized that beta1 and beta3 integrins have partially redundant roles in the adhesion process and that their activation is regulated by similar mechanisms, involving Ca2+-dependent and -independent signaling events and essential functions of talin-1 and kindlin-3 in the terminal activation step.


Subject(s)
Integrins/physiology , Platelet Activation , Blood Platelets/chemistry , Blood Platelets/physiology , Humans , Platelet Membrane Glycoproteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...