Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
Add more filters










Publication year range
1.
J Thromb Haemost ; 16(5): 973-983, 2018 05.
Article in English | MEDLINE | ID: mdl-29488682

ABSTRACT

Essentials Platelet packing density in a hemostatic plug limits molecular movement to diffusion. A diffusion-dependent steep thrombin gradient forms radiating outwards from the injury site. Clot retraction affects the steepness of the gradient by increasing platelet packing density. Together, these effects promote hemostatic plug core formation and inhibit unnecessary growth. SUMMARY: Background Hemostasis studies performed in vivo have shown that hemostatic plugs formed after penetrating injuries are characterized by a core of highly activated, densely packed platelets near the injury site, covered by a shell of less activated and loosely packed platelets. Thrombin production occurs near the injury site, further activating platelets and starting the process of platelet mass retraction. Tightening of interplatelet gaps may then prevent the escape and exchange of solutes. Objectives To reconstruct the hemostatic plug macro- and micro-architecture and examine how platelet mass contraction regulates solute transport and solute concentration in the gaps between platelets. Methods Our approach consisted of three parts. First, platelet aggregates formed in vitro under flow were analyzed using scanning electron microscopy to extract data on porosity and gap size distribution. Second, a three-dimensional (3-D) model was constructed with features matching the platelet aggregates formed in vitro. Finally, the 3-D model was integrated with volume and morphology measurements of hemostatic plugs formed in vivo to determine how solutes move within the platelet plug microenvironment. Results The results show that the hemostatic mass is characterized by extremely narrow gaps, porosity values even smaller than previously estimated and stagnant plasma velocity. Importantly, the concentration of a chemical species released within the platelet mass increases as the gaps between platelets shrink. Conclusions Platelet mass retraction provides a physical mechanism to establish steep chemical concentration gradients that determine the extent of platelet activation and account for the core-and-shell architecture observed in vivo.


Subject(s)
Abdominal Muscles/blood supply , Arterioles/injuries , Blood Platelets/metabolism , Hemostasis , Platelet Aggregation , Thrombin/metabolism , Thrombosis/blood , Vascular System Injuries/blood , Animals , Arterioles/pathology , Arterioles/physiopathology , Blood Flow Velocity , Blood Platelets/pathology , Clot Retraction , Computer Simulation , Diffusion , Disease Models, Animal , Mice, Inbred C57BL , Microcirculation , Models, Biological , Porosity , Thrombosis/pathology , Thrombosis/physiopathology , Time Factors , Vascular System Injuries/pathology , Vascular System Injuries/physiopathology
2.
J Thromb Haemost ; 16(2): 352-363, 2018 02.
Article in English | MEDLINE | ID: mdl-29045015

ABSTRACT

Essentials Signaling by Gas6 through Tyro3/Axl/Mer receptors is essential for stable platelet aggregation. UNC2025 is a small molecule inhibitor of the Mer tyrosine kinase. UNC2025 decreases platelet activation in vitro and thrombus formation in vivo. UNC2025's anti-platelet effect is synergistic with inhibition of the ADP receptor, P2Y12 . SUMMARY: Background Growth arrest-specific protein 6 signals through the TAM (TYRO-3-AXL-MERTK) receptor family, mediating platelet activation and thrombus formation via activation of the aggregate-stabilizing αIIb ß3 integrin. Objective To describe the antithrombotic effects mediated by UNC2025, a small-molecule MERTK tyrosine kinase inhibitor. Methods MERTK phosphorylation and downstream signaling were assessed by immunoblotting. Light transmission aggregometry, flow cytometry and microfluidic analysis were used to evaluate the impact of MERTK inhibition on platelet activation and stability of aggregates in vitro. The effects of MERTK inhibition on arterial and venous thrombosis, platelet accumulation at microvascular injury sites and tail bleeding times were determined with murine models. The effects of combined treatment with ADP-P2Y1&12 pathway antagonists and UNC2025 were also evaluated. Results and Conclusions Treatment with UNC2025 inhibited MERTK phosphorylation and downstream activation of AKT and SRC, decreased platelet activation, and protected animals from pulmonary embolism and arterial thrombosis without increasing bleeding times. The antiplatelet effect of UNC2025 was enhanced in combination with ADP-P2Y1&12 pathway antagonists, and a greater than additive effect was observed when these two agents with different mechanisms of inhibition were coadministered. TAM kinase signaling represents a potential therapeutic target, as inhibition of this axis, especially in combination with ADP-P2Y pathway antagonism, mediates decreased platelet activation, aggregate stability, and thrombus formation, with less hemorrhagic potential than current treatment strategies. The data presented here also demonstrate antithrombotic activity mediated by UNC2025, a novel translational agent, and support the development of TAM kinase inhibitors for clinical applications.


Subject(s)
Adenine/analogs & derivatives , Blood Platelets/drug effects , Piperazines/pharmacology , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Pulmonary Embolism/prevention & control , Thrombosis/prevention & control , c-Mer Tyrosine Kinase/antagonists & inhibitors , Adenine/pharmacokinetics , Adenine/pharmacology , Animals , Blood Platelets/enzymology , Disease Models, Animal , Drug Synergism , Drug Therapy, Combination , Female , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice, Inbred C57BL , Phosphorylation , Piperazines/pharmacokinetics , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins/metabolism , Pulmonary Embolism/blood , Pulmonary Embolism/enzymology , Purinergic P2Y Receptor Antagonists/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Thrombosis/blood , Thrombosis/enzymology , c-Mer Tyrosine Kinase/metabolism , Axl Receptor Tyrosine Kinase
3.
J Thromb Haemost ; 15(3): 526-537, 2017 03.
Article in English | MEDLINE | ID: mdl-27992950

ABSTRACT

Essentials Methods were developed to image the hemostatic response in mouse femoral arteries in real time. Penetrating injuries produced thrombi consisting primarily of platelets. Similar to arterioles, a core-shell architecture of platelet activation occurs in the femoral artery. Differences from arterioles included slower platelet activation and reduced thrombin dependence. SUMMARY: Background Intravital studies performed in the mouse microcirculation show that hemostatic thrombi formed after penetrating injuries develop a characteristic architecture in which a core of fully activated, densely packed platelets is overlaid with a shell of less activated platelets. Objective Large differences in hemodynamics and vessel wall biology distinguish arteries from arterioles. Here we asked whether these differences affect the hemostatic response and alter the impact of anticoagulants and antiplatelet agents. Methods Approaches previously developed for intravital imaging in the mouse microcirculation were adapted to the femoral artery, enabling real-time fluorescence imaging despite the markedly thicker vessel wall. Results Arterial thrombi initiated by penetrating injuries developed the core-and-shell architecture previously observed in the microcirculation. However, although platelet accumulation was greater in arterial thrombi, the kinetics of platelet activation were slower. Inhibiting platelet ADP P2Y12 receptors destabilized the shell and reduced thrombus size without affecting the core. Inhibiting thrombin with hirudin suppressed fibrin accumulation, but had little impact on thrombus size. Removing the platelet collagen receptor, glycoprotein VI, had no effect. Conclusions These results (i) demonstrate the feasibility of performing high-speed fluorescence imaging in larger vessels and (ii) highlight differences as well as similarities in the hemostatic response in the macro- and microcirculation. Similarities include the overall core-and-shell architecture. Differences include the slower kinetics of platelet activation and a smaller contribution from thrombin, which may be due in part to the greater thickness of the arterial wall and the correspondingly greater separation of tissue factor from the vessel lumen.


Subject(s)
Femoral Artery/diagnostic imaging , Hemostasis , Microcirculation , Wounds, Penetrating/therapy , Adenosine Diphosphate/metabolism , Animals , Anticoagulants/pharmacology , Arterioles/metabolism , Blood Coagulation/drug effects , Blood Platelets/metabolism , Femoral Artery/injuries , Fibrin/metabolism , Hemodynamics , Intravital Microscopy , Mice , Mice, Inbred C57BL , Platelet Activation , Platelet Aggregation Inhibitors/pharmacology , Signal Transduction , Thrombin/antagonists & inhibitors , Thrombin/metabolism , Thromboplastin/metabolism , Thrombosis/diagnostic imaging , Thrombosis/drug therapy
4.
Platelets ; 27(7): 673-679, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27809714

ABSTRACT

We have previously shown that Sema4D expressed on the platelet plasma membrane can be cleaved by the metalloprotease ADAM17, producing a 120-kDa exodomain fragment that retains biological activity and remnant fragments of 24-28 kDa that remain associated with the platelet membrane. This process is modulated by calmodulin. Here we investigated the potential role of protein kinase A (PKA) in these events. Using a pharmacological approach, we now show that inhibition of PKA by H89 is sufficient to induce Sema4D exodomain shedding, while activation of PKA inhibits agonist-initiated shedding. Studies on the regulatory mechanism show that the shedding induced by PKA inhibition is mediated by ADAM17, but, unlike agonist-induced shedding, does not involve the dissociation of calmodulin from the Sema4D cytoplasmic domain. In attempt to identify the cleavage sites for shedding, we found that ADAM17 mediates variable cleavages in the juxtamembrane region. Therefore, our data reveal a potential regulatory mechanism for the shedding of Sema4D in platelets.


Subject(s)
Antigens, CD/metabolism , Blood Platelets/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Protein Interaction Domains and Motifs , Semaphorins/metabolism , ADAM17 Protein/metabolism , Antigens, CD/chemistry , Calmodulin/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Activation , Humans , Proteolysis , Semaphorins/chemistry
5.
J Thromb Haemost ; 14(5): 906-17, 2016 05.
Article in English | MEDLINE | ID: mdl-26848552

ABSTRACT

The biophysics of blood flow can dictate the function of molecules and cells in the vasculature with consequent effects on hemostasis, thrombosis, embolism, and fibrinolysis. Flow and transport dynamics are distinct for (i) hemostasis vs. thrombosis and (ii) venous vs. arterial episodes. Intraclot transport changes dramatically the moment hemostasis is achieved or the moment a thrombus becomes fully occlusive. With platelet concentrations that are 50- to 200-fold greater than platelet-rich plasma, clots formed under flow have a different composition and structure compared with blood clotted statically in a tube. The platelet-rich, core/shell architecture is a prominent feature of self-limiting hemostatic clots formed under flow. Importantly, a critical threshold concentration of surface tissue factor is required for fibrin generation under flow. Once initiated by wall-derived tissue factor, thrombin generation and its spatial propagation within a clot can be modulated by γ'-fibrinogen incorporated into fibrin, engageability of activated factor (FIXa)/activated FVIIIa tenase within the clot, platelet-derived polyphosphate, transclot permeation, and reduction of porosity via platelet retraction. Fibrin imparts tremendous strength to a thrombus to resist embolism up to wall shear stresses of 2400 dyne cm(-2) . Extreme flows, as found in severe vessel stenosis or in mechanical assist devices, can cause von Willebrand factor self-association into massive fibers along with shear-induced platelet activation. Pathological von Willebrand factor fibers are A Disintegrin And Metalloprotease with ThromboSpondin-1 domain 13 resistant but are a substrate for fibrin generation due to FXIIa capture. Recently, microfluidic technologies have enhanced the ability to interrogate blood in the context of stenotic flows, acquired von Willebrand disease, hemophilia, traumatic bleeding, and drug action.


Subject(s)
Hemostasis , Rheology , Thrombosis/physiopathology , Animals , Blood Coagulation/drug effects , Blood Flow Velocity , Blood Platelets/drug effects , Constriction, Pathologic , Diffusion , Factor IXa/chemistry , Factor VIIIa/chemistry , Fibrin/chemistry , Fibrinolysis , Humans , Mice , Platelet Activation/drug effects , Platelet Adhesiveness/drug effects , Platelet-Rich Plasma/metabolism , Polyphosphates/chemistry , Porosity , Stress, Mechanical , Thrombin/pharmacology , Thromboplastin/pharmacology , von Willebrand Factor/chemistry
6.
J Thromb Haemost ; 14(5): 1070-81, 2016 05.
Article in English | MEDLINE | ID: mdl-26725377

ABSTRACT

UNLABELLED: Essentials Protein disulfide isomerases may have an essential role in thrombus formation. A platelet-binding sensor (PDI-sAb) was developed to detect thiol reductase activity under flow. Primary human platelet adhesion to collagen at 200 s(-1) was correlated with the PDI-sAb signal. Detected thiol reductase activity was localized in the core of growing thrombi at the site of injury in mice. SUMMARY: Background Protein disulfide isomerases (PDIs) may regulate thrombus formation in vivo, although the sources and targets of PDIs are not fully understood. Methods and results Using click chemistry to link anti-CD61 and a C-terminal azido disulfide-linked peptide construct with a quenched reporter, we developed a fluorogenic platelet-targeting antibody (PDI-sAb) for thiol reductase activity detection in whole blood under flow conditions. PDI-sAb was highly responsive to various exogenous reducing agents (dithiothreitol, glutathione and recombinant PDI) and detected thiol reductase activity on P-selectin/phosphatidylserine-positive platelets activated with convulxin/PAR1 agonist peptide, a signal partially blocked by PDI inhibitors and antibody. In a microfluidic thrombosis model using 4 µg mL(-1) corn trypsin inhibitor-treated human blood perfused over collagen (wall shear rate = 100 s(-1) ), the PDI-sAb signal increased mostly over the first 200 s, whereas platelets continually accumulated for over 500 s, indicating that primary adhesion to collagen, but not secondary aggregation, was correlated with the PDI-sAb signal. Rutin and the PDI blocking antibody RL90 reduced platelet adhesion and the PDI-sAb signal only when thrombin production was inhibited with PPACK, suggesting limited effects of platelet thiol isomerase activity on platelet aggregation on collagen in the presence of thrombin. With anti-mouse CD41 PDI-sAb used in an arteriolar laser injury model, thiol reductase activity was localized in the core of growing thrombi where platelets displayed P-selectin and were in close proximity to disrupted endothelium. Conclusion PDI-sAb is a sensitive and real-time reporter of platelet- and vascular-derived disulfide reduction that targets clots as they form under flow to reveal spatial gradients.


Subject(s)
Blood Platelets/enzymology , Protein Disulfide-Isomerases/metabolism , Sulfhydryl Compounds/chemistry , Animals , Antibodies/chemistry , Blood Flow Velocity , Blood Platelet Disorders/metabolism , Fibrin/chemistry , Hemodynamics , Humans , Integrin beta3/chemistry , Intravital Microscopy , Mice , Microfluidics , Peptides/chemistry , Platelet Adhesiveness , Thrombin/chemistry , Thrombosis/metabolism
8.
J Thromb Haemost ; 10(11): 2344-53, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22978514

ABSTRACT

BACKGROUND: Thrombin undergoes convective and diffusive transport, making it difficult to visualize during thrombosis. We developed the first sensor capable of revealing inner clot thrombin dynamics. METHODS AND RESULTS: An N-terminal-azido thrombin-sensitive fluorescent peptide (ThS-P) with a thrombin-releasable quencher was linked to anti-CD41 using click chemistry to generate a thrombin-sensitive platelet binding sensor (ThS-Ab). Rapid thrombin cleavage of ThS-P (K(m) = 40.3 µm, k(cat) = 1.5 s(-1) ) allowed thrombin monitoring by ThS-P or ThS-Ab in blood treated with 2-25 pm tissue factor (TF). Individual platelets had > 20-fold more ThS-Ab fluorescence after clotting. In a microfluidic assay of whole blood perfusion over collagen ± linked TF (wall shear rate = 100 s(-1) ), ThS-Ab fluorescence increased between 90 and 450 s for 0.1-1 molecule-TF µm(-2) and co-localized with platelets near fibrin. Without TF, neither thrombin nor fibrin was detected on the platelet deposits by 450 s. Using a microfluidic device to control the pressure drop across a thrombus forming on a porous collagen/TF plug (521 s(-1) ), thrombin and fibrin were detected at the thrombus-collagen interface at a zero pressure drop, whereas 80% less thrombin was detected at 3200 Pa in concert with fibrin polymerizing within the collagen. With anti-mouse CD41 ThS-Ab deployed in a mouse laser injury model, the highest levels of thrombin arose between 40 and 160 s nearest the injury site where fibrin co-localized and where the thrombus was most mechanically stable. CONCLUSION: ThS-Ab reveals thrombin locality, which depends on surface TF, flow and intrathrombus pressure gradients.


Subject(s)
Blood Platelets/cytology , Thrombin/chemistry , Animals , Antibodies/chemistry , Blood Coagulation , Collagen/chemistry , Fibrin/chemistry , Hemostasis , Humans , Kinetics , Lasers , Mice , Microfluidic Analytical Techniques , Microfluidics , Peptides/chemistry , Platelet Membrane Glycoprotein IIb/chemistry , Pressure , Protein Transport , Thromboplastin/chemistry , Thrombosis/metabolism , Time Factors
9.
J Thromb Haemost ; 9 Suppl 1: 66-75, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21781243

ABSTRACT

An optimal platelet response to injury can be defined as one in which blood loss is restrained and haemostasis is achieved without the penalty of further tissue damage caused by unwarranted vascular occlusion. This brief review considers some of the ways in which thrombus growth and stability can be regulated so that an optimal platelet response can be achieved in vivo. Three related topics are considered. The first focuses on intracellular mechanisms that regulate the early events of platelet activation downstream of G protein coupled receptors for agonists such as thrombin, thromboxane A(2) and ADP. The second considers the ways in which signalling events that are dependent on stable contacts between platelets can influence the state of platelet activation and thus affect thrombus growth and stability. The third focuses on the changes that are experienced by platelets as they move from their normal environment in freely-flowing plasma to a very different environment within the growing haemostatic plug, an environment in which the narrowing gaps and junctions between platelets not only facilitate communication, but also increasingly limit both the penetration of plasma and the exodus of platelet-derived bioactive molecules.


Subject(s)
Thrombosis/pathology , Vascular Diseases/pathology , Animals , Humans , Mice , Platelet Activation , Signal Transduction , Vascular Diseases/metabolism
12.
J Thromb Haemost ; 7(11): 1886-96, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19740102

ABSTRACT

BACKGROUND: In resting platelets, endothelial cell specific adhesion molecule (ESAM) is located in alpha granules, increasing its cell surface expression following platelet activation. However, the function of ESAM on platelets is unknown. OBJECTIVE: To determine whether ESAM has a role in thrombus formation. METHODS AND RESULTS: We found that following platelet activation ESAM localizes to the junctions between adjacent platelets, suggesting a role for this protein in contact-dependent events that regulate thrombus formation. To test this hypothesis we examined the effect of ESAM deletion on platelet function. In vivo, ESAM(-/-) mice achieved more stable hemostasis than wild-type mice following tail transection, and developed larger thrombi following laser injury of cremaster muscle arterioles. In vitro, ESAM(-/-) platelets aggregated at lower concentrations of G protein-dependent agonists than wild-type platelets, and were more resistant to disaggregation. In contrast, agonist-induced calcium mobilization, alpha(IIb)beta(3) activation, alpha-granule secretion and platelet spreading, were normal in ESAM-deficient platelets. To understand the molecular mechanism by which ESAM regulates platelet activity, we utilized a PDZ domain array to identify the scaffold protein NHERF-1 as an ESAM binding protein, and further demonstrated that it associates with ESAM in both resting and activated platelets. CONCLUSIONS: These findings support a model in which ESAM localizes to platelet contacts following platelet activation in order to limit thrombus growth and stability so that the optimal hemostatic response occurs following vascular injury.


Subject(s)
Cell Adhesion Molecules/metabolism , Intercellular Junctions/chemistry , Platelet Adhesiveness , Thrombosis/pathology , Animals , Hemostasis , Mice , Mice, Knockout , Platelet Activation , Protein Transport , Thrombosis/etiology
13.
J Thromb Haemost ; 6(12): 2193-201, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18983510

ABSTRACT

BACKGROUND: Flow chambers allow the ex vivo study of platelet response to defined surfaces at controlled wall shear stresses. However, most assays require 1-10 mL of blood and are poorly suited for murine whole blood experiments. OBJECTIVE: To measure murine platelet deposition and stability in response to focal zones of prothrombotic stimuli using 100 microL of whole blood and controlled flow exposure. METHODS: Microfluidic methods were used for patterning acid-soluble collagen in 100 microm x 100 microm patches and creating flow channels with a volume of 150 nL. Within 1 min of collection into PPACK and fluorescent anti-mouse CD41 mAb, whole blood from normal mice or from mice deficient in the integrin alpha(2) subunit was perfused for 5 min over the patterned collagen. Platelet accumulation was measured at venous and arterial wall shear rates. After 5 min, thrombus stability was measured with a 'shear step-up' to 8000 s(-1). RESULTS: Wild-type murine platelets adhered and aggregated on collagen in a biphasic shear-dependent manner with increased deposition from 100 to 400 s(-1), but decreased deposition at 1000 s(-1). Adhesion to patterned collagen was severely diminished for platelets lacking a functional alpha(2)beta(1) integrin. Those integrin alpha(2)-deficient platelets that did adhere were removed from the surface when challenged to shear step-up. PAR4 agonist (AYPGKF) treatment of the thrombus at 5 min enhanced aggregate stability during the shear step-up. CONCLUSIONS: PAR4 signaling enhances aggregate stability by mechanisms independent of other thrombin-dependent pathways such as fibrin formation.


Subject(s)
Microfluidics , Platelet Adhesiveness , Receptors, Thrombin/physiology , Thrombosis/pathology , Animals , Collagen/metabolism , Disease Models, Animal , Integrin alpha2beta1 , Mice , Platelet Aggregation , Receptors, Thrombin/agonists , Receptors, Thrombin/metabolism , Signal Transduction , Stress, Mechanical
14.
J Thromb Haemost ; 1(7): 1613-27, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12871298

ABSTRACT

Signaling events downstream from collagen receptors and G protein-coupled receptors are responsible for the initiation and extension of platelet plug formation. This creates the platelet plug and hopefully results in the cessation of bleeding. It is not, however, all that is required for hemostasis, and growing evidence is emerging that the perpetuation of a stable hemostatic plug requires additional intracellular signaling. At least part of this process is made possible by the persistent close contacts between platelets that can only occur after the onset of aggregation. This review discusses several examples of such signaling mechanisms that help to perpetuate the platelet plug in a contact-dependent manner, including outside-in signaling through integrins, signaling though Eph kinases and ephrins, and the role of CD40L.


Subject(s)
Platelet Activation , Signal Transduction , Adenosine Triphosphatases/biosynthesis , Amino Acid Sequence , Animals , Antigens, CD/biosynthesis , Apyrase , Blood Platelets/metabolism , CD40 Ligand/metabolism , Ephrins/metabolism , Glutathione Transferase/metabolism , Humans , Integrins/metabolism , Ligands , Models, Biological , Molecular Sequence Data , Phosphorylation , Protein Structure, Tertiary , Receptor, EphA1/metabolism , Sequence Homology, Amino Acid , Structure-Activity Relationship , Time Factors
15.
Blood ; 97(10): 3109-16, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11342437

ABSTRACT

Cell signaling by coagulation factor Xa (Xa) contributes to pro-inflammatory responses in vivo. This study characterizes the signaling mechanism of Xa in a HeLa cell line that expresses protease-activated receptor 1 (PAR-1) but not PAR-2, -3, or -4. Xa induced NF-kappaB in HeLa cells efficiently but with delayed kinetics compared to thrombin. This delay caused no difference in gene expression patterns, as determined by high-density microarray analysis. Both proteases prominently induced the angiogenesis-promoting gene Cyr61 and connective tissue growth factor. Inhibition of PAR-1 cleavage abolished MAP kinase phosphorylation and gene induction by Xa, demonstrating that Xa signals through PAR-1 and not through a novel member of the PAR family. Activation of cell surface prothrombin with the snake venom enzyme Ecarin also produced PAR-1-dependent signaling. However, though the response to Ecarin was completely blocked by the thrombin inhibitor hirudin, the response to Xa was not. This suggests that the Xa response is not mediated by locally generated thrombin. The concentration dependence of Xa for PAR-1 activation is consistent with previously characterized Xa-mediated PAR-2 signaling, suggesting that local concentration of Xa on the cell surface, rather than sequence-specific recognition of the PAR scissile bond, determines receptor cleavage. This study demonstrates that PAR-1 cleavage by Xa can elicit the same cellular response as thrombin, but mechanistic differences in receptor recognition may be crucial for specific roles for Xa in signaling during spatial or temporal separation from thrombin generation.


Subject(s)
Factor Xa/pharmacology , Gene Expression , Intercellular Signaling Peptides and Proteins , Receptors, Thrombin/genetics , Signal Transduction , Antithrombins/pharmacology , Cell Line , Connective Tissue Growth Factor , Cysteine-Rich Protein 61 , Endothelium, Vascular , Enzyme Activation/drug effects , Growth Substances/genetics , HeLa Cells , Hirudins/pharmacology , Humans , Immediate-Early Proteins/genetics , Kinetics , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Oligonucleotide Array Sequence Analysis , Phosphorylation , Receptor, PAR-1 , Thrombin/metabolism , Thrombin/pharmacology , Umbilical Veins
16.
J Biol Chem ; 276(28): 26276-84, 2001 Jul 13.
Article in English | MEDLINE | ID: mdl-11335723

ABSTRACT

Human pulmonary mast cells (MCs) express tryptases alpha and beta I, and both granule serine proteases are exocytosed during inflammatory events. Recombinant forms of these tryptases were generated for the first time to evaluate their substrate specificities at the biochemical level and then to address their physiologic roles in pulmonary inflammation. Analysis of a tryptase-specific, phage display peptide library revealed that tryptase beta I prefers to cleave peptides with 1 or more Pro residues flanked by 2 positively charged residues. Although recombinant tryptase beta I was unable to activate cultured cells that express different types of protease-activated receptors, the numbers of neutrophils increased >100-fold when enzymatically active tryptase beta I was instilled into the lungs of mice. In contrast, the numbers of lymphocytes and eosinophils in the airspaces did not change significantly. More important, the tryptase beta I-treated mice exhibited normal airway responsiveness. Neutrophils did not extravasate into the lungs of tryptase alpha-treated mice. Thus, this is the first study to demonstrate that the two nearly identical human MC tryptases are functionally distinct in vivo. When MC-deficient W/W(v) mice were given enzymatically active tryptase beta I or its inactive zymogen before pulmonary infection with Klebsiella pneumoniae, tryptase beta I-treated W/W(v) mice had fewer viable bacteria in their lungs relative to zymogen-treated W/W(v) mice. Because neutrophils are required to combat bacterial infections, human tryptase beta I plays a critical role in the antibacterial host defenses of the lung by recruiting neutrophils in a manner that does not alter airway reactivity.


Subject(s)
Lung/enzymology , Mast Cells/enzymology , Pneumonia, Bacterial/enzymology , Serine Endopeptidases/metabolism , Amino Acid Sequence , Animals , Humans , Klebsiella Infections/enzymology , Klebsiella Infections/pathology , Klebsiella pneumoniae/enzymology , Lung/pathology , Mast Cells/pathology , Mice , Molecular Sequence Data , Neutrophils/pathology , Pneumonia, Bacterial/pathology , Substrate Specificity , Tryptases
17.
Oncogene ; 20(13): 1570-81, 2001 Mar 26.
Article in English | MEDLINE | ID: mdl-11313904

ABSTRACT

The four PAR family members are G protein coupled receptors that are normally activated by proteolytic exposure of an occult tethered ligand. Three of the family members are thrombin receptors. The fourth (PAR2) is not activated by thrombin, but can be activated by other proteases, including trypsin, tryptase and Factor Xa. This review focuses on recent information about the manner in which signaling through these receptors is initiated and terminated, including evidence for inter- as well as intramolecular modes of activation, and continuing efforts to identify additional, biologically-relevant proteases that can activate PAR family members.


Subject(s)
Endopeptidases/metabolism , GTP-Binding Proteins/metabolism , Receptors, Thrombin/metabolism , Signal Transduction , Amino Acid Sequence , Binding Sites , Models, Biological , Molecular Sequence Data , Receptor, PAR-2
18.
Thromb Haemost ; 85(3): 533-8, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11307827

ABSTRACT

Three members of the protease-activated receptor family, PAR1, PAR3 and PAR4, are activated when thrombin cleaves the receptor N-terminus, exposing a tethered ligand. Proteases other than thrombin can also cleave PAR family members and, depending upon whether this exposes or removes the tethered ligand, either activate or disable the receptor. For example, on human platelets PAR1 is disabled by cathepsin G, although aggregation still occurs because cathepsin G can activate PAR4. The present studies examine the interaction of cathepsin G and a second neutrophil protease, elastase, with PAR3 using two model systems: COS-7 cells transfected with human PAR3 and mouse platelets, which express PAR3 and PAR4, but not PAR1. In contrast to human platelets, cathepsin G did not aggregate murine platelets, and prevented their activation only at low thrombin concentrations. Elastase had no effect on thrombin responses in mouse platelets, but when added to COS cells expressing human PAR3, both cathepsin G and elastase prevented activation of phospholipase C by thrombin. Notably, this inhibition occurred without loss of the binding sites for two monoclonal antibodies that flank the tethered ligand on human PAR3. We therefore conclude that 1) exposure to cathepsin G disables signaling through human PAR3, and prevents murine PAR3 from serving its normal role, which is to facilitate PAR4 cleavage at low thrombin concentrations, 2) elastase disables human, but not murine, PAR3, 3) in contrast to human PAR4, mouse PAR4 will not support platelet aggregation in response to cathepsin G, and 4) the inactivation of human PAR3 by cathepsin G and elastase involves a mechanism other than amputation of the tethered ligand domain. These results extend the range of possible interactions between PAR family members and proteases, and provide further support for species-specific differences in the interaction of these receptors with proteases other than thrombin.


Subject(s)
Neutrophils/enzymology , Receptors, Thrombin/metabolism , Serine Endopeptidases/metabolism , Amino Acid Sequence , Animals , Blood Platelets/drug effects , COS Cells , Cathepsin G , Cathepsins/metabolism , Humans , Molecular Sequence Data , Pancreatic Elastase/metabolism , Platelet Aggregation/drug effects , Receptors, Thrombin/antagonists & inhibitors , Thrombin/pharmacology , Transfection , Type C Phospholipases/drug effects , Type C Phospholipases/metabolism
19.
Proc Natl Acad Sci U S A ; 97(18): 9984-9, 2000 Aug 29.
Article in English | MEDLINE | ID: mdl-10954748

ABSTRACT

Heterotrimeric G proteins mediate the earliest step in cell responses to external events by linking cell surface receptors to intracellular signaling pathways. G(z) is a member of the G(i) family of G proteins that is prominently expressed in platelets and brain. Here, we show that deletion of the alpha subunit of G(z) in mice: (i) impairs platelet aggregation by preventing the inhibition of cAMP formation normally seen at physiologic concentrations of epinephrine, and (ii) causes the mice to be more resistant to fatal thromboembolism. Loss of G(zalpha) also results in greatly exaggerated responses to cocaine, reduces the analgesic effects of morphine, and abolishes the effects of widely used antidepressant drugs that act as catecholamine reuptake inhibitors. These changes occur despite the presence of other G(ialpha) family members in the same cells and are not accompanied by detectable compensatory changes in the level of expression of other G protein subunits. Therefore, these results provide insights into receptor selectivity among G proteins and a model for understanding platelet function and the effects of psychoactive drugs.


Subject(s)
Blood Platelets/physiology , Brain/physiology , GTP-Binding Protein alpha Subunits , Heterotrimeric GTP-Binding Proteins/metabolism , Platelet Activation/physiology , Psychotropic Drugs/pharmacology , Animals , Blood Platelets/drug effects , Brain/drug effects , Cocaine/pharmacology , Collagen/pharmacology , Cyclic AMP/blood , Epinephrine/pharmacology , Heterotrimeric GTP-Binding Proteins/deficiency , Heterotrimeric GTP-Binding Proteins/genetics , Humans , Lung/drug effects , Lung/physiopathology , Male , Mice , Mice, Knockout , Morphine/pharmacology , Motor Activity , Platelet Aggregation , Signal Transduction , Thromboembolism/genetics , Thromboembolism/physiopathology , Thromboembolism/prevention & control
20.
J Biol Chem ; 275(41): 32129-34, 2000 Oct 13.
Article in English | MEDLINE | ID: mdl-10906137

ABSTRACT

Yeast two-hybrid techniques were used to identify possible effectors for the heterotrimeric G protein G(z) in human bone marrow cells. Eya2, a human homologue of the Drosophila Eya transcription co-activator, was identified. Eya2 interacts with activated Galpha(z) and at least one other member of the Galpha(i) family, Galpha(i2). Interactions were confirmed in mammalian two-hybrid and glutathione S-transferase fusion protein pull-down assays. Regions of Eya2-mediating interaction were mapped to the C-terminal Eya consensus domain. Eya2 is an intrinsically cytosolic protein that is translocated to the nucleus by members of the Six homeodomain-containing family of proteins. Activated Galpha(z) and Galpha(i2) prevent Eya2 translocation and inhibit Six/Eya2-mediated activation of a reporter gene controlled through the MEF3/TATA promoter. Although G proteins are known to regulate the activity of numerous transcription factors, this regulation is normally achieved indirectly via one or more intermediates. We show here a novel functional regulation of a co-activator directly by G protein subunits.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Trans-Activators/metabolism , Animals , Bone Marrow Cells/metabolism , Cell Nucleus/metabolism , Consensus Sequence , Cytosol/metabolism , Fluorescent Antibody Technique , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Genes, Reporter/genetics , HeLa Cells , Heterotrimeric GTP-Binding Proteins/genetics , Humans , Intracellular Signaling Peptides and Proteins , Nerve Tissue Proteins/metabolism , Nuclear Proteins , Promoter Regions, Genetic/genetics , Protein Binding , Protein Subunits , Protein Transport , Protein Tyrosine Phosphatases , Trans-Activators/antagonists & inhibitors , Trans-Activators/chemistry , Trans-Activators/genetics , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...