Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Nat Immunol ; 19(9): 942-953, 2018 09.
Article in English | MEDLINE | ID: mdl-30111894

ABSTRACT

The sensing of microbial genetic material by leukocytes often elicits beneficial pro-inflammatory cytokines, but dysregulated responses can cause severe pathogenesis. Genome-wide association studies have linked the gene encoding phospholipase D3 (PLD3) to Alzheimer's disease and have linked PLD4 to rheumatoid arthritis and systemic sclerosis. PLD3 and PLD4 are endolysosomal proteins whose functions are obscure. Here, PLD4-deficient mice were found to have an inflammatory disease, marked by elevated levels of interferon-γ (IFN-γ) and splenomegaly. These phenotypes were traced to altered responsiveness of PLD4-deficient dendritic cells to ligands of the single-stranded DNA sensor TLR9. Macrophages from PLD3-deficient mice also had exaggerated TLR9 responses. Although PLD4 and PLD3 were presumed to be phospholipases, we found that they are 5' exonucleases, probably identical to spleen phosphodiesterase, that break down TLR9 ligands. Mice deficient in both PLD3 and PLD4 developed lethal liver inflammation in early life, which indicates that both enzymes are needed to regulate inflammatory cytokine responses via the degradation of nucleic acids.


Subject(s)
Dendritic Cells/physiology , Endosomes/metabolism , Exonucleases/metabolism , Hepatitis/genetics , Macrophages/physiology , Membrane Glycoproteins/metabolism , Phospholipase D/metabolism , Alzheimer Disease/genetics , Animals , Arthritis, Rheumatoid/genetics , DNA, Single-Stranded/immunology , Exonucleases/genetics , Genome-Wide Association Study , Humans , Interferon-gamma/metabolism , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phospholipase D/genetics , Scleroderma, Systemic/genetics , Signal Transduction , Toll-Like Receptor 9/metabolism
2.
Proc Natl Acad Sci U S A ; 119(48): e2212659119, 2022 11 29.
Article in English | MEDLINE | ID: mdl-36409883

ABSTRACT

Platelets play a role not only in hemostasis and thrombosis, but also in inflammation and innate immunity. We previously reported that an activated form of tyrosyl-tRNA synthetase (YRSACT) has an extratranslational activity that enhances megakaryopoiesis and platelet production in mice. Here, we report that YRSACT mimics inflammatory stress inducing a unique megakaryocyte (MK) population with stem cell (Sca1) and myeloid (F4/80) markers through a mechanism dependent on Toll-like receptor (TLR) activation and type I interferon (IFN-I) signaling. This mimicry of inflammatory stress by YRSACT was studied in mice infected by lymphocytic choriomeningitis virus (LCMV). Using Sca1/EGFP transgenic mice, we demonstrated that IFN-I induced by YRSACT or LCMV infection suppressed normal hematopoiesis while activating an alternative pathway of thrombopoiesis. Platelets of inflammatory origin (Sca1/EGFP+) were a relevant proportion of those circulating during recovery from thrombocytopenia. Analysis of these "inflammatory" MKs and platelets suggested their origin in myeloid/MK-biased hematopoietic stem cells (HSCs) that bypassed the classical MK-erythroid progenitor (MEP) pathway to replenish platelets and promote recovery from thrombocytopenia. Notably, inflammatory platelets displayed enhanced agonist-induced activation and procoagulant activities. Moreover, myeloid/MK-biased progenitors and MKs were mobilized from the bone marrow, as evidenced by their presence in the lung microvasculature within fibrin-containing microthrombi. Our results define the function of YRSACT in platelet generation and contribute to elucidate platelet alterations in number and function during viral infection.


Subject(s)
Spinocerebellar Ataxias , Thrombocytopenia , Thrombosis , Tyrosine-tRNA Ligase , Virus Diseases , Mice , Animals , Thrombopoiesis , Mice, Transgenic
3.
Haematologica ; 107(9): 2133-2143, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35142156

ABSTRACT

Type 2B von Willebrand disease (VWD) is caused by gain-of-function mutations in von Willebrand factor (VWF). Increased VWF affinity for GPIba results in loss of high molecular weight multimers and enhanced platelet clearance, both contributing to the bleeding phenotype. Severity of the symptoms vary among type 2B VWD patients, with some developing thrombocytopenia only under stress conditions. Efforts have been made to study underlying pathophysiology for platelet abnormalities, but animal studies have been limited because of species specificity in the VWF-GPIba interaction. Here, we generated a severe form of type 2B VWD (p.V1316M) knockin mice in the context of human VWF exon 28 (encoding A1 and A2 domains) and crossed them with human GPIba transgenic strain. Heterozygous mutant mice recapitulated the phenotype of type 2B VWD in autosomal dominant manner and presented severe macrothrombocytopenia. Of note, platelets remaining in the circulation had extracytoplasmic GPIba shed-off from the cell surface. Reciprocal bone marrow transplantation determined mutant VWF produced from endothelial cells as the major cause of the platelet phenotype in type 2B VWD mice. Moreover, altered megakaryocyte maturation in the bone marrow and enhanced extramedullary megakaryopoiesis in the spleen were observed. Interestingly, injection of anti-VWF A1 blocking antibody (NMC-4) not only ameliorated platelet count and GPIba expression, but also reversed MK ploidy shift. In conclusion, we present a type 2B VWD mouse model with humanized VWF-GPIba interaction which demonstrated direct influence of aberrant VWF-GPIba binding on megakaryocytes.


Subject(s)
Thrombocytopenia , von Willebrand Disease, Type 2 , von Willebrand Diseases , Animals , Blood Platelets/metabolism , Endothelial Cells/metabolism , Humans , Mice , Platelet Glycoprotein GPIb-IX Complex/metabolism , Thrombocytopenia/metabolism , von Willebrand Disease, Type 2/genetics , von Willebrand Diseases/metabolism , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
4.
Proc Natl Acad Sci U S A ; 115(33): E7814-E7823, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30061383

ABSTRACT

Understanding of T cell exhaustion and successful therapy to restore T cell function was first described using Clone (Cl) 13 variant selected from the lymphocytic choriomeningitis virus (LCMV) Armstrong (ARM) 53b parental strain. T cell exhaustion plays a pivotal role in both persistent infections and cancers of mice and humans. C57BL/6, BALB, SWR/J, A/J, 129, C3H, and all but one collaborative cross (CC) mouse strain following Cl 13 infection have immunosuppressed T cell responses, high PD-1, and viral titers leading to persistent infection and normal life spans. In contrast, the profile of FVB/N, NZB, PL/J, SL/J, and CC NZO mice challenged with Cl 13 is a robust T cell response, high titers of virus, PD-1, and Lag3 markers on T cells. These mice all die 7 to 9 d after Cl 13 infection. Death is due to enhanced pulmonary endothelial vascular permeability, pulmonary edema, collapse of alveolar air spaces, and respiratory failure. Pathogenesis involves abundant levels of Cl 13 receptor alpha-dystroglycan on endothelial cells, with high viral replication in such cells leading to immunopathologic injury. Death is aborted by blockade of interferon-1 (IFN-1) signaling or deletion of CD8 T cells.


Subject(s)
CD8-Positive T-Lymphocytes , Interferon Type I , Lymphocytic Choriomeningitis , Lymphocytic choriomeningitis virus/physiology , Virus Replication/genetics , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Humans , Interferon Type I/genetics , Interferon Type I/metabolism , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/metabolism , Lymphocytic Choriomeningitis/pathology , Mice , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Lymphocyte Activation Gene 3 Protein
5.
Proc Natl Acad Sci U S A ; 115(35): E8228-E8235, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30104364

ABSTRACT

New mechanisms behind blood cell formation continue to be uncovered, with therapeutic approaches for hematological diseases being of great interest. Here we report an enzyme in protein synthesis, known for cell-based activities beyond translation, is a factor inducing megakaryocyte-biased hematopoiesis, most likely under stress conditions. We show an activated form of tyrosyl-tRNA synthetase (YRSACT), prepared either by rationally designed mutagenesis or alternative splicing, induces expansion of a previously unrecognized high-ploidy Sca-1+ megakaryocyte population capable of accelerating platelet replenishment after depletion. Moreover, YRSACT targets monocytic cells to induce secretion of transacting cytokines that enhance megakaryocyte expansion stimulating the Toll-like receptor/MyD88 pathway. Platelet replenishment by YRSACT is independent of thrombopoietin (TPO), as evidenced by expansion of the megakaryocytes from induced pluripotent stem cell-derived hematopoietic stem cells from a patient deficient in TPO signaling. We suggest megakaryocyte-biased hematopoiesis induced by YRSACT offers new approaches for treating thrombocytopenia, boosting yields from cell-culture production of platelet concentrates for transfusion, and bridging therapy for hematopoietic stem cell transplantation.


Subject(s)
Blood Platelets/metabolism , Hematopoiesis , Megakaryocytes/metabolism , Polyploidy , Thrombocytopenia/metabolism , Tyrosine-tRNA Ligase/metabolism , Blood Platelets/pathology , Cell Culture Techniques , Cells, Cultured , Female , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Male , Megakaryocytes/pathology , Signal Transduction , Thrombocytopenia/pathology , Thrombopoietin/metabolism
6.
Proc Natl Acad Sci U S A ; 111(24): 8925-30, 2014 Jun 17.
Article in English | MEDLINE | ID: mdl-24889626

ABSTRACT

The outcome of a viral infection reflects the balance between virus virulence and host susceptibility. The clone 13 (Cl13) variant of lymphocytic choriomeningitis virus--a prototype of Old World arenaviruses closely related to Lassa fever virus--elicits in C57BL/6 and BALB/c mice abundant negative immunoregulatory molecules, associated with T-cell exhaustion, negligible T-cell-mediated injury, and high virus titers that persist. Conversely, here we report that in NZB mice, despite the efficient induction of immunoregulatory molecules and high viremia, Cl13 generated a robust cytotoxic T-cell response, resulting in thrombocytopenia, pulmonary endothelial cell loss, vascular leakage, and death within 6-8 d. These pathogenic events required type I IFN (IFN-I) signaling on nonhematopoietic cells and were completely abrogated by IFN-I receptor blockade. Thus, IFN-I may play a prominent role in hemorrhagic fevers and other acute virus infections associated with severe vascular pathology, and targeting IFN-I or downstream effector molecules may be an effective therapeutic approach.


Subject(s)
Interferon Type I/metabolism , Lassa Fever/virology , Vascular Diseases/virology , Animals , Bronchoalveolar Lavage , Cell Line , Cricetinae , Cytokines/metabolism , Female , Lassa virus , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NZB , Mice, Transgenic , Signal Transduction , Stem Cells/chemistry , T-Lymphocytes, Cytotoxic/virology , Virus Activation
7.
Blood ; 119(9): 2149-58, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22134168

ABSTRACT

Vascular development and angiogenesis initially depend on endothelial tip cell invasion, which is followed by a series of maturation steps, including lumen formation and recruitment of perivascular cells. Notch ligands expressed on the endothelium and their cognate receptors expressed on perivascular cells are involved in blood vessel maturation, though little is known regarding the Notch-dependent effectors that facilitate perivascular coverage of nascent vessels. Here, we report that vascular smooth muscle cell (VSMC) recognition of the Notch ligand Jagged1 on endothelial cells leads to expression of integrin αvß3 on VSMCs. Once expressed, integrin αvß3 facilitates VSMC adhesion to VWF in the endothelial basement membrane of developing retinal arteries, leading to vessel maturation. Genetic or pharmacologic disruption of Jagged1, Notch, αvß3, or VWF suppresses VSMC coverage of nascent vessels and arterial maturation during vascular development. Therefore, we define a Notch-mediated interaction between the developing endothelium and VSMCs leading to adhesion of VSMCs to the endothelial basement membrane and arterial maturation.


Subject(s)
Basement Membrane/metabolism , Cell Adhesion/physiology , Endothelium, Vascular/metabolism , Integrins/metabolism , Muscle, Smooth, Vascular/metabolism , Receptors, Notch/metabolism , Animals , Arteries/metabolism , Calcium-Binding Proteins/metabolism , Endothelial Cells/metabolism , Gene Expression Regulation , Humans , Integrins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myocytes, Smooth Muscle/metabolism , Neovascularization, Physiologic/genetics , Protein Binding , RNA, Messenger/metabolism , Receptors, Notch/genetics , Serrate-Jagged Proteins , Signal Transduction/physiology , von Willebrand Factor/metabolism
8.
Proc Natl Acad Sci U S A ; 108(21): 8628-33, 2011 May 24.
Article in English | MEDLINE | ID: mdl-21555542

ABSTRACT

The involvement of exosite I in α-thrombin (FIIa) binding to platelet glycoprotein Ibα (GPIbα), which could influence interactions with other substrates, remains undefined. To address the problem, we generated the GPIbα amino terminal domain (GPIbα-N) fully sulfated on three tyrosine residues and solved the structure of its complex with FIIa. We found that sulfotyrosine (Tys) 278 enhances the interaction mainly by establishing contacts with exosite I. We then evaluated how substituting tyrosine with phenylalanine, which cannot be sulfated, affects FIIa binding to soluble or surface-immobilized GPIbα-N. Mutating Tyr(276), which mostly contacts exosite II residues, markedly reduced FIIa interaction with both soluble and immobilized GPIbα-N; mutating Tyr(278) or Tyr(279), which mostly contact exosite I residues, reduced FIIa complexing in solution by 0-20% but affinity for immobilized GPIbα-N 2 to 6-fold, respectively. Moreover, three exosite I ligands--aptamer HD1, hirugen, and lepirudin--did not interfere with soluble FIIa complexing to GPIbα-N, excluding that their binding caused allosteric effects influencing the interaction; nonetheless, all impaired FIIa binding to immobilized GPIbα-N and platelet GPIb nearly as much as aptamer HD22 and heparin, both exosite II ligands. Bound HD1 and hirugen alter Trp(148) orientation in a loop near exosite I preventing contacts with the sulfate oxygen atoms of Tys(279). These results support a mechanism in which binding occurs when the two exosites of one FIIa molecule independently interact with two immobilized GPIbα molecules. Through exosite engagement, GPIbα may influence FIIa-dependent processes relevant to hemostasis and thrombosis.


Subject(s)
Platelet Glycoprotein GPIb-IX Complex/chemistry , Prothrombin/chemistry , Thrombin/chemistry , Tyrosine/analogs & derivatives , Binding Sites , Hemostasis , Humans , Immobilized Proteins , Protein Binding , Protein Structure, Tertiary , Thrombosis , Tyrosine/metabolism
9.
Nat Cardiovasc Res ; 2(4): 368-382, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37206993

ABSTRACT

The activation of platelets and coagulation at vascular injury sites is crucial for haemostasis but can promote thrombosis and inflammation in vascular pathologies. Here, we delineate an unexpected spatio-temporal control mechanism of thrombin activity that is platelet orchestrated and locally limits excessive fibrin formation after initial haemostatic platelet deposition. During platelet activation, the abundant platelet glycoprotein (GP) V is cleaved by thrombin. We demonstrate with genetic and pharmacological approaches that thrombin-mediated shedding of GPV does not primarily regulate platelet activation in thrombus formation, but rather has a distinct function after platelet deposition and specifically limits thrombin-dependent generation of fibrin, a crucial mediator of vascular thrombo-inflammation. Genetic or pharmacologic defects in haemostatic platelet function are unexpectedly attenuated by specific blockade of GPV shedding, indicating that the spatio-temporal control of thrombin-dependent fibrin generation also represents a potential therapeutic target to improve haemostasis.

10.
Cell Physiol Biochem ; 27(2): 139-48, 2011.
Article in English | MEDLINE | ID: mdl-21325831

ABSTRACT

Serotonin (5-hydroxytryptamine, 5-HT) is a neurotransmitter involved in the regulation of numerous neuro-physiological processes. The circulating level of 5-HT is regulated by the membrane transporter SERT present both in the presynaptic nerve terminals and blood platelets. 5-HT transport is a process tightly regulated by a variety of factors including protein phosphorylation. Aim of this study was to ascertain if also the SERT Tyr-phosphorylation mediated by Syk-kinase concurs to the regulation of SERT activity. Indeed we found that 5-HT uptake decreased upon platelet exposure to piceatannol or Syk-inhibitor II, two structurally unrelated inhibitors of the tyrosine-kinase Syk. Tyr-phosphorylation of anti-SERT-immuno-stained proteins in membrane extracts and in anti-SERT-immuno-precipitates, decreased upon platelet treatment with piceatannol, in parallel with a reduction of Syk-activity. Syk was immuno-revealed in the anti-SERT immuno-precipitates, which displayed a piceatannol-sensitive kinase activity towards SERT itself and the Syk-substrate α-sinuclein. Syk inhibitors also caused a decrease of the monensin-induced 5-HT-efflux from platelets and of imipramine binding to them. It is concluded that, in addition to the phosphorylation of SERT mediated by various other kinases, also that catalyzed by Syk might play an important role in the 5-HT transport, likely favoring the transporter conformation exposing the neurotransmitter binding sites.


Subject(s)
Blood Platelets/enzymology , Intracellular Signaling Peptides and Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Serotonin/metabolism , Antidepressive Agents, Tricyclic/chemistry , Antidepressive Agents, Tricyclic/pharmacology , Humans , Imipramine/chemistry , Imipramine/pharmacology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Phosphorylation , Protein Binding , Protein-Tyrosine Kinases/antagonists & inhibitors , Serotonin Plasma Membrane Transport Proteins/metabolism , Stilbenes/pharmacology , Syk Kinase
11.
Blood Adv ; 2(19): 2522-2532, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30287479

ABSTRACT

The interaction of platelet glycoprotein Ibα (GPIbα) with von Willebrand factor (VWF) initiates hemostasis after vascular injury and also contributes to pathological thrombosis. GPIbα binding to the VWF A1 domain (VWFA1) is a target for antithrombotic intervention, but attempts to develop pharmacologic inhibitors have been hindered by the lack of animal models because of the species specificity of the interaction. To address this problem, we generated a knockin mouse with Vwf exon 28-encoding domains A1 and A2 replaced by the human homolog (VWFh28). VWFh28 mice (M1HA) were crossbred with a transgenic mouse strain expressing human GPIbα on platelets (mGPIbαnull;hGPIbαTg; H1MA) to generate a new strain (H1HA) with humanized GPIbα-VWFA1 binding. Plasma VWF levels in the latter 3 strains were similar to those of wild-type mice (M1MA). Compared with the strains that had homospecific GPIbα-VWF pairing (M1MA and H1HA), M1HA mice of those with heterospecific pairing had a markedly greater prolongation of tail bleeding time and attenuation of thrombogenesis after injury to the carotid artery than H1MA mice. Measurements of GPIbα-VWFA1 binding affinity by surface plasmon resonance agreed with the extent of observed functional defects. Ristocetin-induced platelet aggregation was similar in H1HA mouse and human platelet-rich plasma, and it was comparably inhibited by monoclonal antibody NMC-4, which is known to block human GPIbα-VWFA1 binding, which also inhibited FeCl3-induced mouse carotid artery thrombosis. Thus, the H1HA mouse strain is a fully humanized model of platelet GPIbα-VWFA1 binding that provides mechanistic and pharmacologic information relevant to human hemostatic and thrombotic disorders.


Subject(s)
Platelet Glycoprotein GPIb-IX Complex/metabolism , von Willebrand Factor/metabolism , Animals , Biomarkers , Blood Platelets/metabolism , Crosses, Genetic , Exons , Hemostasis , Humans , Mice , Mice, Transgenic , Molecular Docking Simulation , Molecular Dynamics Simulation , Platelet Glycoprotein GPIb-IX Complex/chemistry , Platelet Glycoprotein GPIb-IX Complex/genetics , Protein Aggregates , Protein Binding , Protein Conformation , Protein Multimerization , Structure-Activity Relationship , Surface Plasmon Resonance , Thrombosis/etiology , Thrombosis/metabolism , von Willebrand Factor/chemistry , von Willebrand Factor/genetics
12.
Nat Commun ; 6: 7835, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-26204458

ABSTRACT

Thrombin is a central regulator of leukocyte recruitment and inflammation at sites of vascular injury, a function thought to involve primarily endothelial PAR cleavage. Here we demonstrate the existence of a distinct leukocyte-trafficking mechanism regulated by components of the haemostatic system, including platelet PAR4, GPIbα and fibrin. Utilizing a mouse endothelial injury model we show that thrombin cleavage of platelet PAR4 promotes leukocyte recruitment to sites of vascular injury. This process is negatively regulated by GPIbα, as seen in mice with abrogated thrombin-platelet GPIbα binding (hGPIbα(D277N)). In addition, we demonstrate that fibrin limits leukocyte trafficking by forming a physical barrier to intravascular leukocyte migration. These studies demonstrate a distinct 'checkpoint' mechanism of leukocyte trafficking involving balanced thrombin interactions with PAR4, GPIbα and fibrin. Dysregulation of this checkpoint mechanism is likely to contribute to the development of thromboinflammatory disorders.


Subject(s)
Leukocytes/physiology , Platelet Activation , Platelet Glycoprotein GPIb-IX Complex/metabolism , Receptors, Thrombin/metabolism , Thrombin/metabolism , Animals , Cell Movement , Endothelial Cells/physiology , Fibrinolysis , Humans , Male , Mice , Mice, Inbred C57BL , Models, Animal
13.
J Clin Invest ; 121(11): 4537-47, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22019589

ABSTRACT

Fetal and neonatal immune thrombocytopenia (FNIT) is a severe bleeding disorder caused by maternal antibody-mediated destruction of fetal/neonatal platelets. It is the most common cause of severe thrombocytopenia in neonates, but the frequency of FNIT-related miscarriage is unknown, and the mechanism(s) underlying fetal mortality have not been explored. Furthermore, although platelet αIIbß3 integrin and GPIbα are the major antibody targets in immune thrombocytopenia, the reported incidence of anti-GPIbα-mediated FNIT is rare. Here, we developed mouse models of FNIT mediated by antibodies specific for GPIbα and ß3 integrin and compared their pathogenesis. We found, unexpectedly, that miscarriage occurred in the majority of pregnancies in our model of anti-GPIbα-mediated FNIT, which was far more frequent than in anti-ß3-mediated FNIT. Dams with anti-GPIbα antibodies exhibited extensive fibrin deposition and apoptosis/necrosis in their placentas, which severely impaired placental function. Furthermore, anti-GPIbα (but not anti-ß3) antiserum activated platelets and enhanced fibrin formation in vitro and thrombus formation in vivo. Importantly, treatment with either intravenous IgG or a monoclonal antibody specific for the neonatal Fc receptor efficiently prevented anti-GPIbα-mediated FNIT. Thus, the maternal immune response to fetal GPIbα causes what we believe to be a previously unidentified, nonclassical FNIT (i.e., spontaneous miscarriage but not neonatal bleeding) in mice. These results suggest that a similar pathology may have masked the severity and frequency of human anti-GPIbα-mediated FNIT, but also point to possible therapeutic interventions.


Subject(s)
Abortion, Spontaneous/etiology , Abortion, Spontaneous/prevention & control , Histocompatibility, Maternal-Fetal/immunology , Immunoglobulins, Intravenous/therapeutic use , Platelet Glycoprotein GPIb-IX Complex/immunology , Receptors, Fc/antagonists & inhibitors , Abortion, Spontaneous/immunology , Animals , Blood Platelets/immunology , Disease Models, Animal , Female , Histocompatibility Antigens Class I/immunology , Humans , Integrin beta3/genetics , Integrin beta3/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Platelet Glycoprotein GPIb-IX Complex/genetics , Pregnancy , Receptors, Fc/immunology , Thrombocytopenia, Neonatal Alloimmune/etiology , Thrombocytopenia, Neonatal Alloimmune/immunology
14.
Thromb Haemost ; 104(5): 894-902, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20941453

ABSTRACT

The main question concerning the mechanism of a-thrombin binding to platelet membrane glycoprotein (GP)Ib is whether it involves both thrombin exosite I and exosite II. The solution of two independent crystal structures suggests alternative explanations that may actually reflect different modes of binding with distinct pathophysiological significance. With respect to function, it is still unclear whether thrombin binding to GPIb promotes procoagulant and prothrombotic pathways of response to vascular injury or limits such responses by sequestering, at least temporarily, the active enzyme. We review here published information on these topics and touch upon ongoing studies aimed at finding definitive answers to outstanding questions relevant for a better understanding of thrombosis and haemostasis.


Subject(s)
Hemostasis , Platelet Glycoprotein GPIb-IX Complex/metabolism , Thrombin/metabolism , Thrombosis/blood , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , History, 20th Century , Humans , Models, Molecular , Molecular Sequence Data , Platelet Glycoprotein GPIb-IX Complex/chemistry , Protein Conformation , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Structure-Activity Relationship , Thrombin/chemistry , Thrombosis/history , von Willebrand Factor/metabolism
15.
Cell Physiol Biochem ; 21(1-3): 87-94, 2008.
Article in English | MEDLINE | ID: mdl-18209475

ABSTRACT

BACKGROUND/AIM: platelets possess tightly regulated systems for serotonin (5-HT) transport. This study analysed whether the 5-HT transport mediated by the plasma-membrane transporter SERT is regulated by its Tyr-phosphorylation. METHODS: 5-HT transport was determined by filtration techniques, while immunoblotting procedures were adopted for detecting the Tyr-phosphorylation of SERT in human platelet fractions. RESULTS: 5-HT accumulation in platelets pre-treated with reserpine, which prevents the neurotransmitter transport into the dense granules, decreased upon cellular exposure to PP2 and SU6656, two structurally unrelated inhibitors of Src-kinases. By contrast, the protein Tyr-phosphatase inhibitor pervanadate increased the 5-HT accumulation. Anti-SERT immunostaining of the platelet fractions showed a major band displaying an apparent molecular mass of 50 kappaDa, indicating that, during the analytical procedure, SERT underwent proteolysis, which was counteracted by addition of 4 M urea in the cellular disrupting medium. The Tyr-phosphorylation degree of SERT immunoprecipitated from membrane extracts decreased by platelet treatment with SU6656 or PP2, and enhanced upon pervanadate treatment. The anti-SERT immunoprecipitates displayed anti-Src immunostaining and in vitro kinase activity towards a Src-specific peptide-substrate. Platelet treatment with PP2 or SU6656 also caused a decrease in the imipramine binding to platelets. It was concluded that the Src-mediated SERT Tyr-phosphorylation regulates the 5-HT transport by affecting the neurotransmitter binding sites.


Subject(s)
Blood Platelets/enzymology , Phosphotyrosine/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin/metabolism , Biological Transport/drug effects , Blood Platelets/drug effects , Catalysis/drug effects , Cell Membrane/drug effects , Cell Membrane/enzymology , Humans , Monensin/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins pp60(c-src)/antagonists & inhibitors , Reserpine/pharmacology , Subcellular Fractions/drug effects
16.
Platelets ; 18(5): 357-64, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17654305

ABSTRACT

In the present study some new beta-lactam compounds were screened for their ability to inhibit human platelet activation. In particular four compounds differing in the group on the nitrogen atom of the azetidinone ring were investigated. A beta-lactam having an ethyl 2-carboxyethanoate N-bound group was demonstrated to inhibit, in the micromolar range, both the Ca(2+) release from endoplasmic reticulum, induced either by thrombin or by the ATPase inhibitor thapsigargin, and the Ca(2+) entry in platelets driven by emptying the endoplasmic reticulum. The compound also inhibited the platelet aggregation induced by a variety of physiological agonists including ADP, collagen, thrombin and thrombin mimetic peptide TRAP. The beta-lactam reduced the phosphorylation of pleckstrin (apparent MW 47 kDa), elicited by thrombin but not by the protein kinase C activator phorbol ester. Accordingly it did not significantly affect the aggregation evoked by phorbol ester or Ca(2+) ionophore. It was concluded that the beta-lactam likely exerts its anti-platelet-activating action by hampering the agonist induced cellular Ca(2+) movements. The beta-lactam concentration, which significantly inhibited platelet activation, only negligibly affected the cellular viability. Even if it is still premature to draw definitive conclusions, the present results suggest that this new compound might constitute a tool of potential clinical interest and the starting-point for the synthesis of new more beneficial anti-thrombotic compounds.


Subject(s)
Blood Platelets/metabolism , Monobactams/pharmacology , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , beta-Lactams/pharmacology , Blood Proteins/metabolism , Calcium/metabolism , Carcinogens/pharmacology , Cell Survival/drug effects , Endoplasmic Reticulum/metabolism , Enzyme Inhibitors/pharmacology , Humans , Phorbol Esters/pharmacology , Phosphoproteins/metabolism , Phosphorylation/drug effects , Thapsigargin/pharmacology , Thrombin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL