Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
J Thromb Haemost ; 17(2): 403-414, 2019 02.
Article in English | MEDLINE | ID: mdl-30456926

ABSTRACT

Essentials Neutrophil extracellular traps (NETs) are generated during deep vein thrombosis (DVT). The role of interferon γ (IFNγ) and natural killer (NK) cells in NET formation was studied. IFNγ promote venous thrombosis through NET formation. NK cell depletion reduces DVT. SUMMARY: Background Neutrophils contribute to venous thrombosis through the release of neutrophil extracellular traps (NETs), but the mechanism triggering their formation remains unclear. In vitro data show that interferon (IFN)-γ induces the formation of NETs. Objectives To determine whether IFN-γ and the transcription factor T-box expressed on T cells (Tbet) promote venous thrombosis through neutrophil activation. Methods Venous thrombosis was induced by flow restriction in the inferior vena cava in IFN-γ-/- , Tbet-/- or wild-type (WT) mice. After 48 h, thrombus size was measured by the use of high-frequency ultrasound. NET formation was determined by immunofluorescence. Results and Conclusions Thrombus formation was reduced in Tbet-/- and IFN-γ-/- mice, suggesting that Tbet/IFN-γ-expressing cells are required for venous thrombosis. The number of NETs formed during thrombosis was significantly lower in Tbet-/- and IFN-γ-/- mice. NET formation was also decreased in WT mice treated with an IFN-γ-blocking antibody. Injection of recombinant IFN-γ into IFN-γ-/- mice rescued the phenotype. Natural killer (NK) cells were specifically depleted prior to venous thrombosis induction. NK cell depletion results in decreased NET formation and smaller thrombi, suggesting that NK cells are required for thrombus development. In depleted mice, adoptive transfer of WT NK cells induced a similar thrombosis burden as in WT mice. In contrast, adoptive transfer of IFN-γ -/- NK cells resulted in thrombi similar in size to those in depleted mice. In vitro, we showed that WT neutrophils released fewer NETs when they were cocultured with IFN-γ-/- NK cells. This study demonstrates that NK cell-dependent IFN-γ production is crucial for thrombus development by promoting the formation of NETs by neutrophils.


Subject(s)
Blood Coagulation , Extracellular Traps/metabolism , Killer Cells, Natural/metabolism , Neutrophils/metabolism , Vena Cava, Inferior/metabolism , Venous Thrombosis/metabolism , Animals , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Extracellular Traps/immunology , Interferon-gamma/genetics , Interferon-gamma/metabolism , Killer Cells, Natural/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Signal Transduction , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Vena Cava, Inferior/immunology , Venous Thrombosis/genetics , Venous Thrombosis/immunology
2.
J Thromb Haemost ; 13(12): 2260-72, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26414399

ABSTRACT

BACKGROUND: Growth arrest-specific 6 (Gas6)-deficient mice are protected against venous thromboembolism (VTE), suggesting a role for Gas6 in this disorder. We previously demonstrated that Gas6 induces forkhead box O1 (FoxO-1) phosphorylation through the phosphoinositide 3-kinase-Akt pathway. FoxO-1 regulates the expression of vascular cell adhesion molecule-1 (VCAM-1), a molecule that has been implicated in VTE. OBJECTIVES: To assess the role of FoxO-1 in Gas6-dependent VCAM-1 expression. METHODS: Thrombin was used to stimulate endothelial cells (ECs). Wild-type (WT) and Gas6(-/-) ECs were transfected with small interfering RNA targeting Axl or FoxO-1, a luciferase-coupled plasmid containing the FoxO-1 consensus sequence, and a phosphorylation-resistant FoxO-1 mutant, or treated with an Akt inhibitor. VCAM-1 mRNA expression was measured by real time-qPCR. VCAM-1 protein expression and FoxO-1 and Akt phosphorylation were assessed by western blot analysis. FoxO-1 localization was assessed by immunofluorescence. Adhesion of bone marrow mononuclear cells (BM-MCs) on ECs was assessed by fluorescence. RESULTS AND CONCLUSIONS: Thrombin induces both VCAM-1 expression and FoxO-1 phosphorylation and nuclear exclusion in WT ECs only. Silencing of FoxO-1 enhances VCAM-1 expression in both WT and Gas6(-/-) ECs. Inhibition of Akt or FoxO-1 phosphorylation prevents VCAM-1 expression in WT ECs. These data show that Gas6 induces FoxO-1 phosphorylation, leading to derepression of VCAM-1 expression. BM-MC-EC adhesion is increased by thrombin in WT ECs. BM-MC-EC adhesion is further increased when FoxO-1 is silenced, but decreased when FoxO-1 phosphorylation is inhibited. These results demonstrate that the Gas6-FoxO-1 signaling axis plays an important role in VCAM-1 expression in the context of VTE by promoting BM-MC-EC adhesion.


Subject(s)
Endothelial Cells/drug effects , Forkhead Transcription Factors/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Thrombin/pharmacology , Vascular Cell Adhesion Molecule-1/metabolism , Animals , Bone Marrow Cells/metabolism , Cell Adhesion , Cells, Cultured , Endothelial Cells/metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Genotype , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Mutation , Phenotype , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Signal Transduction , Time Factors , Transfection , Vascular Cell Adhesion Molecule-1/genetics , Venous Thromboembolism/genetics , Venous Thromboembolism/metabolism
3.
J Thromb Haemost ; 12(3): 395-408, 2014.
Article in English | MEDLINE | ID: mdl-24354620

ABSTRACT

BACKGROUND: Gas6 has been shown to interact with Axl in endothelial cells and to induce several signaling pathways involved in cell survival and proliferation. However, the interaction of Gas6/Axl with lipid raft/caveolin-1 in endothelial cells and its role in thrombosis are unknown. OBJECTIVES: We tested whether Axl and/or caveolin-1 is involved in Gas6-induced Akt, ERK1/2, and c-Src activation leading to altered tissue factor expression in endothelial cells. METHODS: Gas6-treated endothelial cells were transfected with small interfering RNA (siRNA) for Axl, caveolin-1, c-Src, and Akt or treated with pharmacological inhibitors of c-Src and ERK1/2. Sucrose gradient centrifugation and confocal microscopy were used to study lipid raft/caveolin-1-enriched fractions. Akt, ERK1/2, p38, and c-Src activation was analyzed by Western blot analysis. Tissue factor expression was assessed by real-time quantitative polymerase chain reaction and immunofluorescence. RESULTS AND CONCLUSION: Gas6 induced Axl and c-Src localization into lipid raft/caveolin-1-enriched fractions. Gas6 increased the phosphorylation of Akt, ERK1/2, and c-Src but not p38. Using siRNA, we demonstrated that Axl is required for Akt, ERK1/2, and c-Src activation after Gas6 stimulation. siRNA for caveolin-1 blocked Gas6-induced phosphorylation of Akt, ERK1/2, and c-Src. c-Src downregulation inhibited Gas6-induced Akt but not ERK1/2 phosphorylation. Finally, Gas6 increased tissue factor mRNA and protein expression in endothelial cells. Tissue factor expression was blocked by siRNA for Axl, caveolin-1, or Akt as well as c-Src inhibition. These data demonstrate that the signaling pathway Gas6/Axl/caveolin-1/c-Src/Akt is required for tissue factor expression in endothelial cells, providing mechanistic insight into how Gas6 exerts its prothrombotic role in the vasculature.


Subject(s)
Caveolin 1/metabolism , Endothelial Cells/cytology , Intercellular Signaling Peptides and Proteins/metabolism , Thromboplastin/metabolism , CSK Tyrosine-Protein Kinase , Cell Proliferation , Cell Survival , Human Umbilical Vein Endothelial Cells , Humans , Membrane Microdomains/chemistry , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Thrombosis/metabolism , src-Family Kinases/metabolism , Axl Receptor Tyrosine Kinase
4.
J Thromb Haemost ; 10(3): 447-52, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22212403

ABSTRACT

BACKGROUND: Venous thrombosis (VT) is an important cause of morbidity and mortality in clinical medicine. Animal models studying venous thrombosis are scarce and, in most cases, very crude and rely on sacrificing the animals to excise formed thrombi. Developing an in vivo murine model of venous thrombosis can be a powerful tool for studying venous thrombosis. OBJECTIVES: We sought to use a high-frequency ultrasound system (HFUS) to dynamically and non-invasively monitor thrombus formation in the inferior vena cava (IVC) of mice. METHODS: We developed a murine model of venous thrombosis using, for detection, the Vevo 770(®), a micro-imaging HFUS. Two different thrombosis models were used to generate thrombi in the IVC of C57Bl/6NCr mice: (i) ligation and (ii) application of ferric chloride (FeCl(3)). We then assessed venous thrombosis by HFUS. RESULTS: In both models, measurements of the clot pathologically correlated favorably with measurements acquired with HFUS. Thrombus develops less than an hour after ligation or FeCl(3) -induced injury of the IVC and the size of the clot increases over time for up to 24 h. Importantly, we demonstrate that HFUS can be used to monitor the effect of an anticoagulant such as dalteparin until complete resolution of the thrombus. CONCLUSIONS: These data show that HFUS assesses venous thrombosis in mice reliably and non-invasively. Developing a murine model of thrombosis using more accurate, and clinically more relevant, techniques such as ultrasonography, is a step towards a better understanding of the pathophysiology of venous thromboembolism.


Subject(s)
Blood Coagulation , Monitoring, Physiologic/methods , Ultrasonography, Doppler, Color , Ultrasonography, Doppler, Pulsed , Vena Cava, Inferior/diagnostic imaging , Venous Thrombosis/diagnostic imaging , Animals , Anticoagulants/pharmacology , Blood Coagulation/drug effects , Chlorides , Dalteparin/pharmacology , Disease Models, Animal , Ferric Compounds , Ligation , Male , Mice , Mice, Inbred C57BL , Time Factors , Vena Cava, Inferior/surgery , Venous Thrombosis/blood , Venous Thrombosis/drug therapy , Venous Thrombosis/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...