Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 12(1): 12523, 2022 07 22.
Article in English | MEDLINE | ID: mdl-35869087

ABSTRACT

Whereas the detection of antiphospholipid autoantibodies (aPL) in COVID-19 is of increasing interest, their role is still unclear. We analyzed a large aPL panel in 157 patients with COVID-19 according to the disease severity. We also investigated a potential association between aPL and extracellular DNA (exDNA, n = 85) or circulating markers of neutrophil extracellular traps (NET) such as citrullinated histones H3 (CitH3, n = 49). A total of 157 sera of patients infected by SARS-CoV-2 were collected. A large aPL panel including lupus anticoagulant, anti-cardiolipin and anti-beta-2 glycoprotein I (IgG, IgM and IgA), anti-phosphatidylethanolamine IgA, anti-prothrombin (IgG and IgM) was retrospectively analyzed according to the disease severity. We found a total aPL prevalence of 54.8% with almost half of the cases having aCL IgG. Within an extended panel of aPL, only aCL IgG were associated with COVID-19 severity. Additionally, severe patients displayed higher CitH3 levels than mild patients. Interestingly, we highlighted a significant association between the levels of aCL IgG and exDNA only in aCL positive patients with severe disease. In conclusion, we showed a significant link between aPL, namely aCL IgG, and circulating exDNA in patients with severe form of COVID-19, that could exacerbate the thrombo-inflammatory state related to disease severity.


Subject(s)
Antiphospholipid Syndrome , COVID-19 , Cell-Free Nucleic Acids , Antibodies, Anticardiolipin , Antibodies, Antiphospholipid , Autoantibodies , Humans , Immunoglobulin A , Immunoglobulin G , Immunoglobulin M , Retrospective Studies , SARS-CoV-2
2.
Blood ; 137(16): 2256-2266, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33556175

ABSTRACT

Genome-wide association studies linked expression of the human neutrophil antigen 3b (HNA-3b) epitope on the Slc44a2 protein with a 30% decreased risk of venous thrombosis (VT) in humans. Slc44a2 is a ubiquitous transmembrane protein identified as a receptor for von Willebrand factor (VWF). To explain the link between Slc44a2 and VT, we wanted to determine how Slc44a2 expressing either HNA-3a or HNA-3b on neutrophils could modulate their adhesion and activation on VWF under flow. Transfected HEK293T cells or neutrophils homozygous for the HNA-3a- or HNA-3b-coding allele were purified from healthy donors and perfused in flow chambers coated with VWF at venous shear rates (100 s-1). HNA-3a expression was required for Slc44a2-mediated neutrophil adhesion to VWF at 100 s-1. This adhesion could occur independently of ß2 integrin and was enhanced when neutrophils were preactivated with lipopolysaccharide. Moreover, specific shear conditions with high neutrophil concentration could act as a "second hit," inducing the formation of neutrophil extracellular traps. Neutrophil mobilization was also measured by intravital microscopy in venules from SLC44A2-knockout and wild-type mice after histamine-induced endothelial degranulation. Mice lacking Slc44a2 showed a massive reduction in neutrophil recruitment in inflamed mesenteric venules. Our results show that Slc44a2/HNA-3a is important for the adhesion and activation of neutrophils in veins under inflammation and when submitted to specific shears. The fact that neutrophils expressing Slc44a2/HNA-3b have a different response on VWF in the conditions tested could thus explain the association between HNA-3b and a reduced risk for VT in humans.


Subject(s)
Isoantigens/metabolism , Membrane Glycoproteins/metabolism , Membrane Transport Proteins/metabolism , Neutrophils/cytology , von Willebrand Factor/metabolism , Animals , Blood Circulation , Cell Adhesion , Cells, Cultured , Extracellular Traps/genetics , Extracellular Traps/metabolism , Gene Expression , HEK293 Cells , Humans , Isoantigens/genetics , Male , Membrane Glycoproteins/genetics , Membrane Transport Proteins/genetics , Mice, Inbred C57BL , Neutrophils/metabolism , Venous Thrombosis/genetics , Venous Thrombosis/metabolism
3.
J Thromb Haemost ; 18(7): 1714-1727, 2020 07.
Article in English | MEDLINE | ID: mdl-32297475

ABSTRACT

BACKGROUND: Genome wide association studies (GWAS) identified SLC44A2 as a novel susceptibility gene for venous thrombosis (VT) and previous work established that SLC44A2 contributed to clot formation upon vascular injury. OBJECTIVE: To further investigate the role of SLC44A2 in VT by utilizing SLC44A2 deficient mice (Slc44a2-/- ) in two representative disease models. METHODS: Mice were included in a hypercoagulability model driven by siRNA-mediated hepatic gene silencing of anticoagulants Serpinc1 (antithrombin) and Proc (protein C) and a flow restriction (stenosis) model induced by partial ligation of the inferior vena cava. RESULTS: In the hypercoagulability model, no effect in onset was observed in Slc44a2-/- animals; however, a drop in plasma fibrinogen and von Willebrand factor coinciding with an increase in blood neutrophils was recorded. In the neutrophil dependent stenosis model after 48 hours, Slc44a2-/- mice had significantly smaller thrombi both in length and weight with less platelet accumulation as a percentage of the total thrombus area. During the initiation of thrombosis at 6 hours post-stenosis, Slc44a2-/- mice also had smaller thrombi both in length and weight, with circulating platelets remaining elevated in Slc44a2-/- animals. Platelet activation and aggregation under both static- and venous and arterial shear conditions were normal for blood from Slc44a2-/- mice. CONCLUSIONS: These studies corroborate the original GWAS findings and establish a contributing role for SLC44A2 during the initiation of VT, with indications that this may be related to platelet-neutrophil interaction. The precise mechanism however remains elusive and warrants further investigation.


Subject(s)
Thrombophilia , Venous Thrombosis , Animals , Blood Platelets , Constriction, Pathologic , Disease Models, Animal , Genome-Wide Association Study , Membrane Transport Proteins/genetics , Mice , Platelet Activation , Thrombophilia/genetics , Venous Thrombosis/genetics
4.
Blood ; 120(10): 2133-43, 2012 Sep 06.
Article in English | MEDLINE | ID: mdl-22837532

ABSTRACT

For a long time, blood coagulation and innate immunity have been viewed as interrelated responses. Recently, the presence of leukocytes at the sites of vessel injury has been described. Here we analyzed interaction of neutrophils, monocytes, and platelets in thrombus formation after a laser-induced injury in vivo. Neutrophils immediately adhered to injured vessels, preceding platelets, by binding to the activated endothelium via leukocyte function antigen-1-ICAM-1 interactions. Monocytes rolled on a thrombus 3 to 5 minutes postinjury. The kinetics of thrombus formation and fibrin generation were drastically reduced in low tissue factor (TF) mice whereas the absence of factor XII had no effect. In vitro, TF was detected in neutrophils. In vivo, the inhibition of neutrophil binding to the vessel wall reduced the presence of TF and diminished the generation of fibrin and platelet accumulation. Injection of wild-type neutrophils into low TF mice partially restored the activation of the blood coagulation cascade and accumulation of platelets. Our results show that the interaction of neutrophils with endothelial cells is a critical step preceding platelet accumulation for initiating arterial thrombosis in injured vessels. Targeting neutrophils interacting with endothelial cells may constitute an efficient strategy to reduce thrombosis.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Neutrophils/metabolism , Thromboplastin/genetics , Thrombosis/metabolism , Animals , Blood Coagulation , Blood Platelets/cytology , Blood Vessels/injuries , Blood Vessels/metabolism , Blood Vessels/pathology , Cell Adhesion , Cell Communication , Cell Count , Cell Movement , Endothelial Cells/pathology , Endothelium, Vascular/pathology , Factor XII/metabolism , Factor XII Deficiency/genetics , Factor XII Deficiency/metabolism , Fibrin/metabolism , Intercellular Adhesion Molecule-1/metabolism , Lasers , Mice , Monocytes/cytology , Neutrophils/cytology , Neutrophils/transplantation , Platelet Count , Thromboplastin/deficiency
5.
Thromb Haemost ; 108(3): 527-32, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22782575

ABSTRACT

Recombinant tissue plasminogen activator (r-tPA) is the drug of choice for thrombolysis, but it is associated with a significant risk of bleeding and is not always successful. By cleaving von Willebrand factor (VWF), the metalloprotease ADAMTS13 (a disintegrin-like and metalloprotease with thrombospondin type I repeats-13) down-regulates thrombus formation in injured vessels. We investigated whether recombinant ADAMTS13 (r-ADAMTS13) induces thrombolysis in vivo in mice. Thrombosis was produced by ferric chloride-induced (FeCl(3)) injury in the venules of a dorsal skinfold chamber. Phosphate-buffered saline (PBS, vehicle), r-tPA or r-ADAMTS13, supplemented with hirudin (to stop on-going thrombin generation), was directly applied onto the occluded vessel, and thrombus dissolution was evaluated by intravital microscopy. The incidence of blood flow restoration significantly increased 30 minutes (min) after r-ADAMTS13 vs. PBS treatment (60% vs. 0%, p<0.05) and 60 min after r-tPA treatment (75% vs. 17%, p<0.05). Both r-tPA and r-ADAMTS13 significantly reduced thrombus size 60 min after their superfusion (53.2% and 62.3% of the initial thrombus size, p<0.05 and p<0.01, respectively). Bleeding occurred in all r-tPA-treated chambers, while it was absent in mice treated with r-ADAMTS13 or PBS. We observed that, similar to r-tPA, r-ADAMTS13 can dissolve occlusive thrombi induced by FeCl(3) injury in venules. In contrast to r-tPA, the in vivo thrombolytic effect of ADAMTS13 was not associated with any signs of haemorrhage. ADAMTS13 could represent a new therapeutic option for thrombolysis.


Subject(s)
ADAM Proteins/therapeutic use , Fibrinolytic Agents/therapeutic use , Thrombolytic Therapy , Venous Thrombosis/drug therapy , ADAM Proteins/pharmacology , ADAM Proteins/toxicity , ADAMTS13 Protein , Animals , Chlorides/toxicity , Computer Systems , Disease Models, Animal , Drug Evaluation, Preclinical , Ferric Compounds/toxicity , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/toxicity , Hemorrhage/chemically induced , Humans , Male , Mice , Mice, Inbred C57BL , Microcirculation/drug effects , Microscopy/methods , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Recombinant Proteins/toxicity , Skin Window Technique , Thrombolytic Therapy/adverse effects , Tissue Plasminogen Activator/pharmacology , Tissue Plasminogen Activator/therapeutic use , Venous Thrombosis/chemically induced , Venules
6.
Blood ; 119(26): 6335-43, 2012 Jun 28.
Article in English | MEDLINE | ID: mdl-22596262

ABSTRACT

Transfusion-related acute lung injury (TRALI) is the leading cause of transfusion-related death. The biologic processes contributing to TRALI are poorly understood. All blood products can cause TRALI, and no specific treatment is available. A "2-event model" has been proposed as the trigger. The first event may include surgery, trauma, or infection; the second involves the transfusion of antileukocyte antibodies or bioactive lipids within the blood product. Together, these events induce neutrophil activation in the lungs, causing endothelial damage and capillary leakage. Neutrophils, in response to pathogens or under stress, can release their chromatin coated with granule contents, thus forming neutrophil extracellular traps (NETs). Although protective against infection, these NETs are injurious to tissue. Here we show that NET biomarkers are present in TRALI patients' blood and that NETs are produced in vitro by primed human neutrophils when challenged with anti-HNA-3a antibodies previously implicated in TRALI. NETs are found in alveoli of mice experiencing antibody-mediated TRALI. DNase 1 inhalation prevents their alveolar accumulation and improves arterial oxygen saturation even when administered 90 minutes after TRALI onset. We suggest that NETs form in the lungs during TRALI, contribute to the disease process, and thus could be targeted to prevent or treat TRALI.


Subject(s)
Acute Lung Injury/etiology , DNA/immunology , DNA/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Transfusion Reaction , Acute Lung Injury/immunology , Animals , Blood Donors , Cells, Cultured , Extracellular Space/genetics , Humans , Male , Mice , Mice, Inbred BALB C , Neutrophil Activation/immunology , Neutrophils/pathology , Transplantation Immunology , Transplantation, Homologous/immunology
7.
J Exp Med ; 206(9): 1913-27, 2009 Aug 31.
Article in English | MEDLINE | ID: mdl-19667060

ABSTRACT

Recent publications have demonstrated the presence of tissue factor (TF)-bearing microparticles (MPs) in the blood of patients suffering from cancer. However, whether these MPs are involved in thrombosis remains unknown. We show that pancreatic and lung cancer cells produce MPs that express active TF and P-selectin glycoprotein ligand 1 (PSGL-1). Cancer cell-derived MPs aggregate platelets via a TF-dependent pathway. In vivo, cancer cell-derived MPs, but not their parent cells, infused into a living mouse accumulate at the site of injury and reduce tail bleeding time and the time to occlusion of venules and arterioles. This thrombotic state is also observed in mice developing tumors. In such mice, the amount of circulating platelet-, endothelial cell-, and cancer cell-derived MPs is increased. Endogenous cancer cell-derived MPs shed from the growing tumor are able to accumulate at the site of injury. Infusion of a blocking P-selectin antibody abolishes the thrombotic state observed after injection of MPs or in mice developing a tumor. Collectively, our results indicate that cancer cell-derived MPs bearing PSGL-1 and TF play a key role in thrombus formation in vivo. Targeting these MPs could be of clinical interest in the prevention of thrombosis and to limit formation of metastasis in cancer patients.


Subject(s)
Cell-Derived Microparticles/metabolism , Membrane Glycoproteins/metabolism , Platelet Aggregation/physiology , Thromboplastin/metabolism , Thrombosis/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Humans , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence
8.
J Clin Invest ; 117(12): 3708-19, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18037996

ABSTRACT

Bile salt-dependent lipase (BSDL) is an enzyme involved in the duodenal hydrolysis and absorption of cholesteryl esters. Although some BSDL is transported to blood, the role of circulating BSDL is unknown. Here, we demonstrate that BSDL is stored in platelets and released upon platelet activation. Because BSDL contains a region that is structurally homologous to the V3 loop of HIV-1, which binds to CXC chemokine receptor 4 (CXCR4), we hypothesized that BSDL might bind to CXCR4 present on platelets. In human platelets in vitro, both BSDL and a peptide corresponding to its V3-like loop induced calcium mobilization and enhanced thrombin-mediated platelet aggregation, spreading, and activated alpha(IIb)beta(3) levels. These effects were abolished by CXCR4 inhibition. BSDL also increased the production of prostacyclin by human endothelial cells. In a mouse thrombosis model, BSDL accumulated at sites of vessel wall injury. When CXCR4 was antagonized, the accumulation of BSDL was inhibited and thrombus size was reduced. In BSDL(-/-) mice, calcium mobilization in platelets and thrombus formation were attenuated and tail bleeding times were increased in comparison with those of wild-type mice. We conclude that BSDL plays a role in optimal platelet activation and thrombus formation by interacting with CXCR4 on platelets.


Subject(s)
Blood Platelets/metabolism , Platelet Aggregation , Receptors, CXCR4/metabolism , Sterol Esterase/metabolism , Thrombosis/enzymology , Absorption , Animals , Bleeding Time , Calcium/metabolism , Calcium Signaling/drug effects , Calcium Signaling/genetics , Cell Line , Cholesterol Esters/metabolism , Disease Models, Animal , Duodenum/enzymology , Endothelial Cells/metabolism , Epoprostenol/biosynthesis , Epoprostenol/genetics , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/metabolism , HIV-1/genetics , HIV-1/metabolism , Mice , Mice, Knockout , Platelet Aggregation/drug effects , Platelet Aggregation/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Structure, Secondary , Receptors, CXCR4/agonists , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/genetics , Sequence Homology, Amino Acid , Sterol Esterase/genetics , Sterol Esterase/pharmacology , Thrombosis/chemically induced , Thrombosis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...