Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
Int J Mol Sci ; 22(23)2021 Nov 26.
Article in English | MEDLINE | ID: mdl-34884600

ABSTRACT

A huge effort has been devoted to developing drugs targeting integrins over 30 years, because of the primary roles of integrins in the cell-matrix milieu. Five αv-containing integrins, in the 24 family members, have been a central target of fibrosis. Currently, a small molecule against αvß1 is undergoing a clinical trial for NASH-associated fibrosis as a rare agent aiming at fibrogenesis. Latent TGFß activation, a distinct talent of αv-integrins, has been intriguing as a therapeutic target. None of the αv-integrin inhibitors, however, has been in the clinical market. αv-integrins commonly recognize an Arg-Gly-Asp (RGD) sequence, and thus the pharmacophore of inhibitors for the 5-integrins is based on the same RGD structure. The RGD preference of the integrins, at the same time, dilutes ligand specificity, as the 5-integrins share ligands containing RGD sequence such as fibronectin. With the inherent little specificity in both drugs and targets, "disease specificity" has become less important for the inhibitors than blocking as many αv-integrins. In fact, an almighty inhibitor for αv-integrins, pan-αv, was in a clinical trial. On the contrary, approved integrin inhibitors are all specific to target integrins, which are expressed in a cell-type specific manner: αIIbß3 on platelets, α4ß1, α4ß7 and αLß2 on leukocytes. Herein, "disease specific" integrins would serve as attractive targets. α8ß1 and α11ß1 are selectively expressed in hepatic stellate cells (HSCs) and distinctively induced upon culture activation. The exceptional specificity to activated HSCs reflects a rather "pathology specific" nature of these new integrins. The monoclonal antibodies against α8ß1 and α11ß1 in preclinical examinations may illuminate the road to the first medical agents.


Subject(s)
Hepatic Stellate Cells/drug effects , Integrins/antagonists & inhibitors , Liver Cirrhosis/drug therapy , Receptors, Collagen/antagonists & inhibitors , Animals , Hepatic Stellate Cells/pathology , Humans , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology
2.
Sci Rep ; 7: 40002, 2017 01 05.
Article in English | MEDLINE | ID: mdl-28054640

ABSTRACT

Honokiol, derived from Magnolia officinalis, has various pharmacological properties. Platelet activation plays a critical role in cardiovascular diseases. Honokiol has been reported to inhibit collagen-stimulated rabbit platelet aggregation. However, detailed further studies on the characteristics and functional activity of honokiol in platelet activation are relatively lacking. In the present study, honokiol specifically inhibited platelet aggregation and Ca+2 ion mobilization stimulated with collagen or convulxin, an agonist of glycoprotein (GP) VI, but not with aggretin, an agonist of integrin α2ß1. Honokiol also attenuated the phosphorylation of Lyn, PLCγ2, PKC, MAPKs, and Akt after convulxin stimulation. Honokiol have no cytotoxicity in zebrafish embryos. Honokiol diminished the binding of anti-GP VI (FITC-JAQ1) mAb to human platelets, and it also reduced the coimmunoprecipitation of GP VI-bound Lyn after convulxin stimulation. The surface plasmon resonance results revealed that honokiol binds directly to GP VI, with a KD of 289 µM. Platelet function analysis revealed that honokiol substantially prolonged the closure time in human whole blood and increased the occlusion time of thrombotic platelet plug formation in mice. In conclusion, honokiol acts as a potent antagonist of collagen GP VI in human platelets, and it has therapeutic potential in the prevention of the pathological thrombosis.


Subject(s)
Biphenyl Compounds/metabolism , Lignans/metabolism , Platelet Aggregation/drug effects , Receptors, Collagen/antagonists & inhibitors , Animals , Biphenyl Compounds/toxicity , Humans , Lignans/toxicity , Mice , Protein Binding , Surface Plasmon Resonance , Thrombosis/prevention & control , Zebrafish
3.
J Mol Biol ; 426(13): 2457-70, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24768818

ABSTRACT

The discoidin domain receptors (DDRs), DDR1 and DDR2, form a unique subfamily of receptor tyrosine kinases that are activated by the binding of triple-helical collagen. Excessive signaling by DDR1 and DDR2 has been linked to the progression of various human diseases, including fibrosis, atherosclerosis and cancer. We report the inhibition of these unusual receptor tyrosine kinases by the multi-targeted cancer drugs imatinib and ponatinib, as well as the selective type II inhibitor DDR1-IN-1. Ponatinib is identified as the more potent molecule, which inhibits DDR1 and DDR2 with an IC50 of 9nM. Co-crystal structures of human DDR1 reveal a DFG-out conformation (DFG, Asp-Phe-Gly) of the kinase domain that is stabilized by an unusual salt bridge between the activation loop and αD helix. Differences to Abelson kinase (ABL) are observed in the DDR1 P-loop, where a ß-hairpin replaces the cage-like structure of ABL. P-loop residues in DDR1 that confer drug resistance in ABL are therefore accommodated outside the ATP pocket. Whereas imatinib and ponatinib bind potently to both the DDR and ABL kinases, the hydrophobic interactions of the ABL P-loop appear poorly satisfied by DDR1-IN-1 suggesting a structural basis for its DDR1 selectivity. Such inhibitors may have applications in clinical indications of DDR1 and DDR2 overexpression or mutation, including lung cancer.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Collagen/antagonists & inhibitors , Receptors, Mitogen/antagonists & inhibitors , Amino Acid Sequence , Benzamides/pharmacology , Binding Sites , Discoidin Domain Receptor 1 , Discoidin Domain Receptors , Humans , Imatinib Mesylate , Imidazoles/pharmacology , Models, Molecular , Molecular Sequence Data , Piperazines/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Structure, Secondary , Protein Structure, Tertiary , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/chemistry , Proto-Oncogene Proteins c-abl/genetics , Pyridazines/pharmacology , Pyrimidines/pharmacology , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Collagen/chemistry , Receptors, Collagen/genetics , Receptors, Mitogen/chemistry , Receptors, Mitogen/genetics , Sequence Homology, Amino Acid
4.
J Immunol ; 191(12): 5941-50, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24244022

ABSTRACT

Th17 cells play a critical role in the pathogenesis of rheumatoid arthritis (RA), but the mechanisms by which these cells regulate the development of RA are not fully understood. We have recently shown that α2ß1 integrin, the receptor of type I collagen, is the major collagen-binding integrin expressed by human Th17 cells. In this study, we examined the role of α2ß1 integrin in Th17-mediated destructive arthritis in the murine model of collagen-induced arthritis (CIA). We found that α2ß1 integrin is expressed on synovial Th17 cells from CIA mice and its neutralization with a specific mAb significantly reduced inflammation and cartilage degradation, and protected the mice from bone erosion. Blockade of α2ß1 integrin led to a decrease in the number of Th17 cells in the joints and to a reduction of IL-17 levels in CIA mice. This was associated with an inhibition of receptor activator of NF-κB ligand levels and osteoclast numbers, and reduction of bone loss. We further show that α2ß1 integrin is expressed on synovial Th17 cells from RA patients, and that its ligation with collagen costimulated the production of IL-17 by polarized human Th17 cells by enhancing the expression of retinoic acid receptor-related orphan receptor C through ERK and PI3K/AKT. Our findings provide the first evidence, to our knowledge, that α2ß1 integrin is an important pathway in Th17 cell activation in the pathogenesis of CIA, suggesting that its blockade can be beneficial for the treatment of RA and other Th17-associated autoimmune diseases.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Arthritis, Experimental/therapy , Arthritis, Rheumatoid/metabolism , Integrin alpha2beta1/physiology , Osteolysis/prevention & control , Receptors, Collagen/physiology , Th17 Cells/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibody Specificity , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/immunology , Cartilage, Articular/pathology , Collagen/pharmacology , Cricetinae , Down-Regulation , Female , Humans , Inflammation , Integrin alpha2beta1/antagonists & inhibitors , Interleukin-17/blood , Lymphocyte Activation , MAP Kinase Signaling System , Mice , Mice, Inbred DBA , NF-kappa B/physiology , Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Osteoclasts/pathology , Osteolysis/etiology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , RANK Ligand/blood , Receptors, Collagen/antagonists & inhibitors , Signal Transduction , Synovial Membrane/metabolism , Synovial Membrane/pathology , Th17 Cells/physiology
5.
Cell Death Dis ; 2: e186, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21796158

ABSTRACT

Although mesenchymal stem cells (MSCs) are the natural source for bone regeneration, the exact mechanisms governing MSC crosstalk with collagen I have not yet been uncovered. Cell adhesion to collagen I is mostly mediated by three integrin receptors - α1ß1, α2ß1 and α11ß1. Using human MSC (hMSC), we show that α11 subunit exhibited the highest basal expression levels but on osteogenic stimulation, both α2 and α11 integrins were significantly upregulated. To elucidate the possible roles of collagen-binding integrins, we applied short hairpin RNA (shRNA)-mediated knockdown in hMSC and found that α2 or α11 deficiency, but not α1, results in a tremendous reduction of hMSC numbers owing to mitochondrial leakage accompanied by Bcl-2-associated X protein upregulation. In order to clarify the signaling conveyed by the collagen-binding integrins in hMSC, we analyzed the activation of focal adhesion kinase, extracellular signal-regulated protein kinase and serine/threonine protein kinase B (PKB/Akt) kinases and detected significantly reduced Akt phosphorylation only in α2- and α11-shRNA hMSC. Finally, experiments with hMSC from osteoporotic patients revealed a significant downregulation of α2 integrin concomitant with an augmented mitochondrial permeability. In conclusion, our study describes for the first time that disturbance of α2ß1- or α11ß1-mediated interactions to collagen I results in the cell death of MSCs and urges for further investigations examining the impact of MSCs in bone conditions with abnormal collagen I.


Subject(s)
Collagen/metabolism , Integrin alpha2beta1/metabolism , Integrins/metabolism , Mesenchymal Stem Cells/cytology , Receptors, Collagen/metabolism , Cell Adhesion , Cell Differentiation , Humans , Integrin alpha2beta1/antagonists & inhibitors , Integrin alpha2beta1/genetics , Integrins/antagonists & inhibitors , Integrins/genetics , Mesenchymal Stem Cells/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Collagen/antagonists & inhibitors , Receptors, Collagen/genetics , Up-Regulation , bcl-2-Associated X Protein/metabolism
6.
J Biol Chem ; 285(14): 10434-45, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20129924

ABSTRACT

Fibrotic tissue is characterized by an overabundance of myofibroblasts. Thus, understanding the factors that induce myofibroblast differentiation is paramount to preventing fibrotic healing. Previous studies have shown that mechanical stress derived from the integrin-mediated interaction between extracellular matrix and the cytoskeleton promotes myofibroblast differentiation. Integrin alpha11beta1 is a collagen receptor on fibroblasts. To determine whether alpha11beta1 can act as a mechanosensor to promote the myofibroblast phenotype, mouse embryonic fibroblasts and human corneal fibroblasts were utilized. We found that alpha11 mRNA and protein levels were up-regulated in mouse embryonic fibroblasts grown in attached three-dimensional collagen gels and conversely down-regulated in cells grown in floating gels. alpha11 up-regulation could be prevented by manually detaching the collagen gels or by cytochalasin D treatment. Furthermore, SB-431542, an inhibitor of signaling via ALK4, ALK5, and ALK7, prevented the up-regulation of alpha11 and the concomitant phosphorylation of Smad3 under attached conditions. In attached gels, TGF-beta1 was secreted in its inactive form but surprisingly not further activated, thus not influencing alpha11 regulation. However, inhibition of activin A attenuated the up-regulation of alpha11. To determine the role of alpha11 in myofibroblast differentiation, human corneal fibroblasts were transfected with small interfering RNA to alpha11, which decreased alpha-smooth muscle actin expression and myofibroblast differentiation. Our data suggest that alpha11beta1 is regulated by cell/matrix stress involving activin A and Smad3 and that alpha11beta1 regulates myofibroblast differentiation.


Subject(s)
Activins/metabolism , Cell Differentiation , Cornea/cytology , Fibroblasts/cytology , Integrins/metabolism , Muscle, Skeletal/cytology , Receptors, Collagen/metabolism , Actins/genetics , Actins/metabolism , Activins/genetics , Animals , Blotting, Western , Cell Culture Techniques , Cells, Cultured , Collagen/metabolism , Cornea/metabolism , Cytoskeleton/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/metabolism , Fluorescent Antibody Technique , Humans , Integrins/antagonists & inhibitors , Integrins/genetics , Mice , Muscle, Skeletal/metabolism , Nodal Signaling Ligands/genetics , Nodal Signaling Ligands/metabolism , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Receptors, Collagen/antagonists & inhibitors , Receptors, Collagen/genetics , Reverse Transcriptase Polymerase Chain Reaction , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
8.
Thromb Res ; 122(6): 786-803, 2008.
Article in English | MEDLINE | ID: mdl-18045662

ABSTRACT

Collagen, one of the major proteins of sub-endothelial vasculature get exposed following endothelium denudement, is a potent stimulator of platelet adhesion and aggregation. Adhesion of platelets following endothelial injury is the primary event usually associated with uncontrolled platelet activation culminating into intravascular thrombosis, thus needs to be intervened to prevent the pathology related to various peripheral, myocardial and cerebral ischemic episodes. Recent advances in the understanding of collagen mediated platelet adhesion and aggregation have led to the identification of two prominent receptors, glycoprotein Ia/IIa (GPIa/IIa or integrin alpha(2)beta(1)) and glycoprotein VI (GPVI) and associated intracellular signaling, which are undoubtedly the new emerging targets for the development of more effective antithrombotic drugs. The optimism for collagen antagonism is based on results obtained so far by the use of monoclonal and polyclonal antibodies, peptide inhibitors, knockouts models and collagen-mimetics in various in vitro test systems and animal models. These findings have revealed that collagen receptor inhibition is an attractive and secure strategy for the new drug development to prevent intravascular thrombosis.


Subject(s)
Integrin alpha2/physiology , Platelet Membrane Glycoproteins/physiology , Receptors, Collagen/physiology , Signal Transduction/physiology , Thrombosis/prevention & control , Animals , Calcium/metabolism , Collagen/pharmacology , Humans , Platelet Activation , Platelet Adhesiveness , Polymorphism, Genetic , Receptors, Collagen/antagonists & inhibitors , Receptors, Collagen/genetics
9.
Curr Pharm Des ; 13(26): 2673-83, 2007.
Article in English | MEDLINE | ID: mdl-17897011

ABSTRACT

Platelets have important roles in atherosclerosis and thrombosis and their inhibition reduces the risk of these disorders. There is still a need for platelet inhibitors affecting pathways that reduce thrombosis and atherosclerosis while leaving normal hemostasis relatively unaffected, thus reducing possible bleeding complications. Although combinations show progress in achieving these goals none of the present inhibitors completely fulfill these requirements. Collagen receptors offer attractive possibilities as alternative targets at early stages in platelet activation. Three major collagen receptors are assessed in this review; the alpha2beta1 integrin, responsible primarily for platelet adhesion to collagen; GPVI, the major signaling receptor for collagen; and GPIb-V-IX, which is indirectly a collagen receptor via von Willebrand factor. Several thrombosis models and experimental approaches suggest that all three are interesting targets and merit further investigation.


Subject(s)
Blood Platelets/drug effects , Blood Platelets/metabolism , Drug Delivery Systems , Receptors, Collagen/metabolism , Animals , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Drug Delivery Systems/methods , Humans , Platelet Activation/drug effects , Platelet Activation/physiology , Platelet Aggregation Inhibitors/administration & dosage , Platelet Membrane Glycoproteins/agonists , Platelet Membrane Glycoproteins/metabolism , Receptors, Collagen/agonists , Receptors, Collagen/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology
10.
Biochemistry ; 42(43): 12497-502, 2003 Nov 04.
Article in English | MEDLINE | ID: mdl-14580195

ABSTRACT

Snake venoms contain a number of hemostatically active C-type lectin-like proteins (CLPs), which affect the blood coagulation system, endothelial cells, and platelets. CLPs have broad similarities in structure and possess distinct biological functions. EMS16, a CLP from Echis multisquamatus venom, which is a potent and selective inhibitor of the collagen receptor, glycoprotein Ia/IIa (integrin alpha2beta1), has been used in the present study to examine structure-function relationships in venom CLPs by X-ray crystallography. The structure of EMS16, determined at a resolution of 1.9 A, revealed a heterodimer involved with domain swapping of the central loop as observed in the structures of other CLPs. A part of the glycan was observed and identified as N-acetyl-D-glucosamine (GlcNAc) in the electron density map at Asn21 of subunit B, an expected glycosylation site. EMS16 had a unique, positively charged electrostatic potential patch on the concave surface that may qualify as a site for interaction with the I-domain of the glycoprotein Ia/IIa.


Subject(s)
Lectins, C-Type/chemistry , Receptors, Collagen/antagonists & inhibitors , Viper Venoms/chemistry , Amino Acid Sequence , Animals , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Homology, Amino Acid , Viperidae
11.
J Biochem ; 134(1): 19-23, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12944366

ABSTRACT

EMS16 is a member of the snake venom-derived C-type lectin family of proteins (CLPs) found in the venom of Echis multisquamatus. It binds to glycoprotein Ia/IIa (integrin alpha2beta1), a major collagen receptor of platelets, acting as a potent antagonist of platelet aggregation and cell migration. Amino acid sequencing and cDNA cloning of EMS16 have revealed that it is composed of an A chain of 134 amino acid residues and a B chain of 128 residues. Crystals of EMS16 belong to space group P2(1)2(1)2(1), with unit-cell parameters a = 46.57, b = 59.93, and c = 115.74 A, and diffract to a resolution of 1.9 A. Phase determination is underway by means of molecular replacement with the structure of blood coagulation factor IX-binding protein (IX-bp) from habu snake venom (PDB code 1bj3) as the search model.


Subject(s)
Lectins, C-Type/chemistry , Receptors, Collagen/antagonists & inhibitors , Viper Venoms/chemistry , Amino Acid Sequence , Animals , Base Sequence , Crystallization , Crystallography, X-Ray , DNA, Complementary/genetics , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Molecular Sequence Data , N-Glycosyl Hydrolases/metabolism , Protein Conformation , Sequence Alignment , Sequence Homology, Amino Acid , Viper Venoms/genetics , Viper Venoms/metabolism , Viperidae
12.
Acta Biochim Pol ; 50(4): 1119-28, 2003.
Article in English | MEDLINE | ID: mdl-14739999

ABSTRACT

Direct interactions between collagen, the most thrombogenic component of the extracellular matrix, and platelet surface membrane receptors mediate platelet adhesion and induce platelet activation and aggregation. In this process two glycoproteins are crucial: integrin alpha2beta1, an adhesive receptor, and GPVI, which is especially responsible for signal transduction. Specific antagonists of the collagen receptors are useful tools for investigating the complexity of platelet-collagen interactions. In this work we assessed the usefulness of DGEA peptide (Asp-Gly-Glu-Ala), the shortest collagen type I-derived motif recognised by the collagen-binding integrin alpha2beta1, as a potential antagonist of collagen receptors. We examined platelet function using several methods including platelet adhesion under static conditions, platelet function analyser PFA-100TM, whole blood electric impedance aggregometry (WBEA) and flow cytometry. We found that DGEA significantly inhibited adhesion, aggregation and release reaction of collagen activated blood platelets. The inhibitory effect of DGEA on static platelet adhesion reached sub-maximal values at millimolar inhibitor concentrations, whereas the specific blocker of alpha2beta1 - monoclonal antibodies Gi9, when used at saturating concentrations, had only a moderate inhibitory effect on platelet adhesion. Considering that 25-30% of total collagen binding to alpha2beta1 is specific, we conclude that DGEA is a strong antagonist interfering with a variety of collagen-platelet interactions, and it can be recognised not only by the primary platelet adhesion receptor alpha2beta1 but also by other collagen receptors.


Subject(s)
Blood Platelets/metabolism , Collagen/metabolism , Oligopeptides/metabolism , Adult , Antibodies, Monoclonal/immunology , Female , Humans , Male , Receptors, Collagen/antagonists & inhibitors , Receptors, Collagen/immunology , Receptors, Collagen/metabolism
13.
Thromb Res ; 105(2): 153-60, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11958806

ABSTRACT

Trimucytin is a powerful platelet aggregation inducer isolated from the venom of Taiwan habu snake (Trimeresurus mucrosquamatus). In this study, we found that the snake venom protein, crovidisin, which prevents collagen-platelet interaction through its high-affinity binding to collagen, inhibited competitively trimucytin-induced aggregation of washed human platelets with a pA(2) value of 6.65. The ability of trimucytin in triggering platelet aggregation was suppressed by a monoclonal antibody (A2-IIE10) raised against the alpha2 subunit of alpha2beta1 integrin (glycoprotein Ia/IIa), indicating that platelet alpha2beta1 integrin plays a central role in trimucytin's platelet reactivity. Many studies have localized the major reactive site of alpha2beta1 integrin to the I-domain of alpha2 subunit. However, Escherichia coli-produced recombinant alpha2 I-domain (GST-alpha2 fusion protein) blocking collagen-induced platelet aggregation failed to inhibit aggregation of platelets in response to trimucytin. Based on these findings, it is concluded that the platelet reactivity of trimucytin is alpha2beta1 integrin-dependent, while the I-domain present in the alpha2 subunit is not involved. This novel snake venom protein would be useful for mapping the functional domain of alpha2beta1 integrin.


Subject(s)
Crotalid Venoms/pharmacology , Metalloproteases , Platelet Activation/drug effects , Proteoglycans/pharmacology , Reptilian Proteins , Animals , Antibodies, Monoclonal/pharmacology , Binding Sites , Binding, Competitive , Blood Platelets/chemistry , Blood Platelets/drug effects , Blood Platelets/metabolism , Carrier Proteins/pharmacology , Collagen Type I/antagonists & inhibitors , Collagen Type I/pharmacology , Humans , Integrin alpha2/immunology , Integrin alpha2/metabolism , Integrin alpha2/physiology , Protein Structure, Tertiary , Receptors, Collagen/antagonists & inhibitors , Receptors, Collagen/physiology , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/physiology
14.
Blood Coagul Fibrinolysis ; 13(2): 95-103, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11914651

ABSTRACT

Activation of the prothrombinase complex, which catalyzes the formation of thrombin from prothrombin, is crucial for the (patho)physiological processes of hemostasis and thrombosis. We here report that washed platelets supplemented with prothrombin can be irreversibly aggregated with otherwise non-aggregant doses of adenosine diphosphate (10 micromol/l), thrombin (0.06 U/ml), or collagen (1 microg/ml). Prothrombinase-catalyzed prothrombin to thrombin conversion most probably supports this aggregation response, since inhibitors of thrombin (hirudin or heparin) and an inhibitor of activated factor X (DX-9065a) impair the response. A certain degree of agonist-induced platelet activation seems to be required for this prothrombin-supported aggregation response, since prothrombin alone does not induce aggregation, and blockade of glycoprotein Ia/IIa with a specific antibody inhibits the platelet aggregation response to collagen and prothrombin. These results may suggest that activation of the prothrombinase complex could be a common step of the platelet response to distinct agonists, which may be achieved at low levels of platelet stimulation.


Subject(s)
Platelet Aggregation/drug effects , Thromboplastin/metabolism , Adenosine Diphosphate/pharmacology , Animals , Cattle , Collagen/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Enzyme Activation/drug effects , Factor X/pharmacology , Factor Xa/pharmacology , Humans , Integrin alpha2beta1 , Prothrombin/metabolism , Prothrombin/pharmacology , Receptors, Collagen/antagonists & inhibitors , Thrombin/pharmacology , Thromboplastin/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL