Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Cytokine ; 111: 563-566, 2018 11.
Article in English | MEDLINE | ID: mdl-29807687

ABSTRACT

Endothelial cells (EC) are critical sites of human cytomegalovirus (hCMV) infection in vivo. Infection can induce the production of various EC cytokines, such as interleukin (IL-)6, which can have autocrine and/or paracrine effector functions. Here, we report that hCMV induces the production of EC IL-11, a relatively understudied member of the IL-6-type cytokine family. We detail temporal EC IL-11 translation and protein secretion dynamics in response to hCMV infection, and reveal distinct differences compared to EC IL-6. Viral replication had markedly opposing effects on the regulation of these closely related cytokines, representing a major driving force behind IL-11 production, whilst concurrently suppressing IL-6 expression. This is the first report of any biological agent that stimulates EC IL-11 production.


Subject(s)
Cytomegalovirus Infections/metabolism , Cytomegalovirus/genetics , Endothelial Cells/metabolism , Endothelial Cells/virology , Interleukin-11/metabolism , Virus Replication/genetics , Cells, Cultured , Cytokines/metabolism , Cytomegalovirus Infections/virology , Human Umbilical Vein Endothelial Cells , Humans
2.
Thromb Haemost ; 118(2): 229-250, 2018 02.
Article in English | MEDLINE | ID: mdl-29378352

ABSTRACT

Atherothrombosis is a leading cause of cardiovascular mortality and long-term morbidity. Platelets and coagulation proteases, interacting with circulating cells and in different vascular beds, modify several complex pathologies including atherosclerosis. In the second Maastricht Consensus Conference on Thrombosis, this theme was addressed by diverse scientists from bench to bedside. All presentations were discussed with audience members and the results of these discussions were incorporated in the final document that presents a state-of-the-art reflection of expert opinions and consensus recommendations regarding the following five topics: 1. Risk factors, biomarkers and plaque instability: In atherothrombosis research, more focus on the contribution of specific risk factors like ectopic fat needs to be considered; definitions of atherothrombosis are important distinguishing different phases of disease, including plaque (in)stability; proteomic and metabolomics data are to be added to genetic information. 2. Circulating cells including platelets and atherothrombosis: Mechanisms of leukocyte and macrophage plasticity, migration, and transformation in murine atherosclerosis need to be considered; disease mechanism-based biomarkers need to be identified; experimental systems are needed that incorporate whole-blood flow to understand how red blood cells influence thrombus formation and stability; knowledge on platelet heterogeneity and priming conditions needs to be translated toward the in vivo situation. 3. Coagulation proteases, fibrin(ogen) and thrombus formation: The role of factor (F) XI in thrombosis including the lower margins of this factor related to safe and effective antithrombotic therapy needs to be established; FXI is a key regulator in linking platelets, thrombin generation, and inflammatory mechanisms in a renin-angiotensin dependent manner; however, the impact on thrombin-dependent PAR signaling needs further study; the fundamental mechanisms in FXIII biology and biochemistry and its impact on thrombus biophysical characteristics need to be explored; the interactions of red cells and fibrin formation and its consequences for thrombus formation and lysis need to be addressed. Platelet-fibrin interactions are pivotal determinants of clot formation and stability with potential therapeutic consequences. 4. Preventive and acute treatment of atherothrombosis and arterial embolism; novel ways and tailoring? The role of protease-activated receptor (PAR)-4 vis à vis PAR-1 as target for antithrombotic therapy merits study; ongoing trials on platelet function test-based antiplatelet therapy adjustment support development of practically feasible tests; risk scores for patients with atrial fibrillation need refinement, taking new biomarkers including coagulation into account; risk scores that consider organ system differences in bleeding may have added value; all forms of oral anticoagulant treatment require better organization, including education and emergency access; laboratory testing still needs rapidly available sensitive tests with short turnaround time. 5. Pleiotropy of coagulation proteases, thrombus resolution and ischaemia-reperfusion: Biobanks specifically for thrombus storage and analysis are needed; further studies on novel modified activated protein C-based agents are required including its cytoprotective properties; new avenues for optimizing treatment of patients with ischaemic stroke are needed, also including novel agents that modify fibrinolytic activity (aimed at plasminogen activator inhibitor-1 and thrombin activatable fibrinolysis inhibitor.


Subject(s)
Thromboembolism/therapy , Thrombosis/blood , Thrombosis/therapy , Anticoagulants/therapeutic use , Biomarkers/blood , Blood Coagulation , Erythrocytes/metabolism , Factor VIII/metabolism , Factor XII/metabolism , Factor XIII/metabolism , Humans , Macrophages/metabolism , Netherlands , Phenotype , Plaque, Atherosclerotic/blood , Plaque, Atherosclerotic/diagnosis , Plaque, Atherosclerotic/therapy , Polyphosphates/metabolism , Risk Factors , Signal Transduction , Thromboembolism/blood , Thromboembolism/diagnosis , Thrombosis/diagnosis
3.
J Thromb Haemost ; 14(5): 995-1007, 2016 05.
Article in English | MEDLINE | ID: mdl-26613657

ABSTRACT

UNLABELLED: Essentials How the Alzheimer's disease (AD) peptide ß-amyloid (Aß) disrupts neuronal function in the disease is unclear. Factor (F) XII initiates blood clotting via FXI, and thrombosis has been implicated in AD. Aß triggers FXII-dependent FXI and thrombin activation, evidence of which is seen in AD plasma. Aß-triggered clotting could contribute to neuronal dysfunction in AD and be a novel therapeutic target. SUMMARY: Background ß-Amyloid (Aß) is a key pathologic element in Alzheimer's disease (AD), but the mechanisms by which it disrupts neuronal function in vivo are not completely understood. AD is characterized by a prothrombotic state, which could contribute to neuronal dysfunction by affecting cerebral blood flow and inducing inflammation. The plasma protein factor XII triggers clot formation via the intrinsic coagulation cascade, and has been implicated in thrombosis. Objectives To investigate the potential for Aß to contribute to a prothrombotic state. Methods and results We show that Aß activates FXII, resulting in FXI activation and thrombin generation in human plasma, thereby establishing Aß as a possible driver of prothrombotic states. We provide evidence for this process in AD by demonstrating decreased levels of FXI and its inhibitor C1 esterase inhibitor in AD patient plasma, suggesting chronic activation, inhibition and clearance of FXI in AD. Activation of the intrinsic coagulation pathway in AD is further supported by elevated fibrin levels in AD patient plasma. Conclusions The ability of Aß to promote coagulation via the FXII-driven contact system identifies new mechanisms by which it could contribute to neuronal dysfunction and suggests potential new therapeutic targets in AD.


Subject(s)
Alzheimer Disease/blood , Amyloid beta-Peptides/chemistry , Factor XII/chemistry , Thrombin/chemistry , Amyloid beta-Protein Precursor/chemistry , Animals , Blood Coagulation , Blood Flow Velocity , Cerebrovascular Circulation , Complement C1 Inhibitor Protein/chemistry , Enzyme-Linked Immunosorbent Assay , Female , Fibrin/chemistry , Healthy Volunteers , Humans , Inflammation , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Thrombosis/blood , Treatment Outcome
4.
J Thromb Haemost ; 14(3): 427-37, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26707513

ABSTRACT

The contact system is a plasma protease cascade initiated by factor XII (FXII) that activates the proinflammatory kallikrein-kinin system and the procoagulant intrinsic coagulation pathway. Anionic surfaces induce FXII zymogen activation to form proteolytically active FXIIa. Bacterial surfaces also have the ability to activate contact system proteins, indicating an important role for host defense using the cooperation of the inflammatory and coagulation pathways. Recent research has shown that inorganic polyphosphate found in platelets activates FXII in vivo and can induce coagulation in pathological thrombus formation. Experimental studies have shown that interference with FXII provides thromboprotection without a therapy-associated increase in bleeding, renewing interest in the FXIIa-driven intrinsic pathway of coagulation as a therapeutic target. This review summarizes how the contact system acts as the cross-road of inflammation, coagulation, and innate immunity.


Subject(s)
Blood Coagulation , Factor XII/metabolism , Immunity, Innate , Inflammation Mediators/blood , Inflammation Mediators/immunology , Inflammation/blood , Inflammation/immunology , Angioedemas, Hereditary/blood , Angioedemas, Hereditary/immunology , Animals , Hepatocytes/immunology , Hepatocytes/metabolism , Humans , Hypersensitivity/blood , Hypersensitivity/immunology , Polyphosphates/blood , Signal Transduction , Thrombosis/blood , Thrombosis/immunology
5.
J Intern Med ; 278(6): 571-85, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26373901

ABSTRACT

Plasma protein factor XII (FXII) activates the procoagulant and proinflammatory contact system that drives both the kallikrein-kinin system and the intrinsic pathway of coagulation. When zymogen FXII comes into contact with negatively charged surfaces, it auto-activates to the serine proteaseactivated FXII (FXIIa). Recently, various in vivo activators of FXII have been identified including heparin, misfolded protein aggregates, polyphosphate and nucleic acids. Murine models have established a central role of FXII in arterial and venous thrombosis. Despite its central function in thrombosis, deficiency in FXII does not impair haemostasis in animals and humans. In a preclinical cardiopulmonary bypass system in large animals, the FXIIa-blocking antibody 3F7 prevented thrombosis; however, in contrast to traditional anticoagulants, bleeding was not increased. In addition to its function in thrombosis, FXIIa initiates formation of the inflammatory mediator bradykinin. This mediator increases vascular leak, causes vasodilation, and induces chemotaxis with implications for septic, anaphylactic and allergic disease states. Therefore, targeting FXIIa appears to be a promising strategy for thromboprotection without associated bleeding risks but with anti-inflammatory properties.


Subject(s)
Anticoagulants/pharmacology , Blood Coagulation/drug effects , Factor XIIa/metabolism , Hemorrhage/prevention & control , Inflammation/prevention & control , Thrombosis , Animals , Blood Coagulation/physiology , Drug Discovery , Hemorrhage/chemically induced , Humans , Inflammation/blood , Thrombosis/blood , Thrombosis/physiopathology , Thrombosis/prevention & control
7.
Hamostaseologie ; 35(4): 338-50, 2015.
Article in English | MEDLINE | ID: mdl-25609114

ABSTRACT

Blood coagulation is essential for hemostasis, however excessive coagulation can lead to thrombosis. Factor XII starts the intrinsic coagulation pathway and contact-induced factor XII activation provides the mechanistic basis for the diagnostic aPTT clotting assay. Despite its function for fibrin formation in test tubes, patients and animals lacking factor XII have a completely normal hemostasis. The lack of a bleeding tendency observed in factor XII deficiency states is in sharp contrast to deficiencies of other components of the coagulation cascade and factor XII has been considered to have no function for coagulation in vivo. Recently, experimental animal models showed that factor XII is activated by an inorganic polymer, polyphosphate, which is released from procoagulant platelets and that polyphosphate-driven factor XII activation has an essential role in pathologic thrombus formation. Cumulatively, the data suggest to target polyphosphate, factor XII, or its activated form factor XIIa for anticoagulation. As the factor XII pathway specifically contributes to thrombosis but not to hemostasis, interference with this pathway provides a unique opportunity for safe anticoagulation that is not associated with excess bleeding. The review summarizes current knowledge on factor XII functions, activators and inhibitors.


Subject(s)
Anticoagulants/administration & dosage , Blood Coagulation/immunology , Factor XII/immunology , Factor XIIa/immunology , Thrombosis/immunology , Thrombosis/prevention & control , Animals , Blood Coagulation/drug effects , Factor XII/drug effects , Factor XIIa/drug effects , Humans , Models, Cardiovascular , Models, Immunological , Thrombolytic Therapy/methods , Thrombolytic Therapy/trends
8.
J Thromb Haemost ; 13(5): 732-42, 2015 May.
Article in English | MEDLINE | ID: mdl-25418346

ABSTRACT

BACKGROUND: Acute thrombotic microangiopathies (TMAs) are characterized by excessive microvascular thrombosis and are associated with markers of neutrophil extracellular traps (NETs) in plasma. NETs are composed of DNA fibers and promote thrombus formation through the activation of platelets and clotting factors. OBJECTIVE: The efficient removal of NETs may be required to prevent excessive thrombosis such as in TMAs. To test this hypothesis, we investigated whether TMAs are associated with a defect in the degradation of NETs. METHODS AND RESULTS: We show that NETs generated in vitro were efficiently degraded by plasma from healthy donors. However, NETs remained stable after exposure to plasma from TMA patients. The inability to degrade NETs was linked to a reduced DNase activity in TMA plasma. Plasma DNase1 was required for efficient NET degradation and TMA plasma showed decreased levels of this enzyme. Supplementation of TMA plasma with recombinant human DNase1 restored NET-degradation activity. CONCLUSIONS: Our data indicate that DNase1-mediated degradation of NETs is impaired in patients with TMAs. The role of plasma DNases in thrombosis is, as of yet, poorly understood. Reduced plasma DNase1 activity may cause the persistence of pro-thrombotic NETs and thus promote microvascular thrombosis in TMA patients.


Subject(s)
Deoxyribonuclease I/metabolism , Extracellular Traps/metabolism , Neutrophils/metabolism , Thrombotic Microangiopathies/blood , Humans , Hydrolysis
9.
Thromb Haemost ; 112(1): 53-64, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24671506

ABSTRACT

Heparin-induced thrombocytopenia (HIT) is the most frequent drug-induced immune reaction affecting blood cells. Its antigen is formed when the chemokine platelet factor 4 (PF4) complexes with polyanions. By assessing polyanions of varying length and degree of sulfation using immunoassay and circular dichroism (CD)-spectroscopy, we show that PF4 structural changes resulting in antiparallel ß-sheet content >30% make PF4/polyanion complexes antigenic. Further, we found that polyphosphates (polyP-55) induce antigenic changes on PF4, whereas fondaparinux does not. We provide a model suggesting that conformational changes exposing antigens on PF4/polyanion complexes occur in the hairpin involving AA 32-38, which form together with C-terminal AA (66-70) of the adjacent PF4 monomer a continuous patch on the PF4 tetramer surface, explaining why only tetrameric PF4 molecules express "HIT antigens". The correlation of antibody binding in immunoassays with PF4 structural changes provides the intriguing possibility that CD-spectroscopy could become the first antibody-independent, in vitro method to predict potential immunogenicity of drugs. CD-spectroscopy could identify compounds during preclinical drug development that induce PF4 structural changes correlated with antigenicity. The clinical relevance can then be specifically addressed during clinical trials. Whether these findings can be transferred to other endogenous proteins requires further studies.


Subject(s)
Antigens/immunology , Heparin/metabolism , Platelet Factor 4/metabolism , Polymers/metabolism , Thrombocytopenia/metabolism , Antibodies/immunology , Antigen-Antibody Complex/metabolism , Circular Dichroism , Fondaparinux , Humans , Platelet Factor 4/immunology , Polyelectrolytes , Polyphosphates/metabolism , Polysaccharides/metabolism , Protein Binding , Protein Multimerization , Protein Structure, Secondary , Sulfuric Acid Esters/metabolism , Thrombocytopenia/chemically induced , Thrombocytopenia/immunology
11.
Thromb Haemost ; 110(3): 399-407, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23846131

ABSTRACT

Plasma prekallikrein is the liver-derived precursor of the trypsin-like serine protease plasma kallikrein (PK) and circulates in plasma bound to high molecular weight kininogen. The zymogen is converted to PK by activated factor XII. PK drives multiple proteolytic reaction cascades in the cardiovascular system such as the intrinsic pathway of coagulation, the kallikrein-kinin system, the fibrinolytic system, the renin-angiotensin system and the alternative complement pathway. Here, we review the biochemistry and cell biology of PK and focus on recent in vivo studies that have established important functions of the protease in procoagulant and proinflammatory disease states. Targeting PK offers novel strategies not previously appreciated to interfere with thrombosis and vascular inflammation in a broad variety of diseases.


Subject(s)
Bradykinin/metabolism , Plasma Kallikrein/metabolism , Animals , Aprotinin/chemistry , Blood Coagulation , Cerebral Hemorrhage/metabolism , Complement System Proteins , Cysteine/chemistry , Diabetic Retinopathy/metabolism , Disulfides/chemistry , Factor XIIa/chemistry , Fibrinolysis , Hemostasis , Humans , Inflammation , Kallikrein-Kinin System , Kallikreins/chemistry , Kinins/chemistry , Mice , Oligonucleotides, Antisense/chemistry , Peptides/chemistry , Protein Structure, Tertiary , Proteolysis , Renin-Angiotensin System , Signal Transduction , Thrombosis/metabolism , Trypsin/chemistry
12.
Atherosclerosis ; 226(1): 67-73, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23137827

ABSTRACT

OBJECTIVE: Proliferation of smooth muscle cells (SMCs) can stabilize atherosclerotic lesions but the molecular mechanisms that regulate this process in humans are largely unknown. We have previously shown that heparan sulfate proteoglycans (HSPGs), such as perlecan, regulate SMC growth in animal models by modulating heparin-binding mitogens. Since perlecan is expressed at low levels in human atherosclerosis, we speculated that the effect of heparan sulfate (HS) in human disease was rather influenced by HS degradation and investigated the expression of heparanase (HPSE) in human carotid endarterectomies. METHODS AND RESULTS: Gene expression analysis from 127 endarterectomies in the BiKE database revealed increased expression of HPSE in carotid plaques compared with normal arteries, and a further elevation in symptomatic lesions. Increased HPSE protein expression in symptomatic plaque tissue was verified by tissue microarrays. HPSE mRNA levels correlated positively with expression of inflammatory markers IL-18, RANTES and IL-1ß, and also T-cell co-stimulatory molecules, such as B7.2, CD28, LFA-1 and 4-1BB. Previously reported single nucleotide polymorphisms within HPSE were associated with differential mRNA expression in plaques. Immunohistochemistry revealed that inflammatory cells were major producers of HPSE in plaque tissue. HPSE immunoreactivity was also observed in SMCs adjacent to the necrotic core and was co-localized to deposits of fibrin. CONCLUSIONS: This study demonstrates increased expression of HPSE in human atherosclerosis associated with inflammation, coagulation and plaque instability. Since HS can regulate SMC proliferation and influence plaque stability, the findings suggest that HPSE degradation of HS take part in the regulation of SMC function in human atherosclerosis.


Subject(s)
Carotid Artery Diseases/enzymology , Glucuronidase/biosynthesis , Adult , Carotid Artery Diseases/genetics , Gene Expression Regulation , Glucuronidase/genetics , Humans , Middle Aged
13.
J Thromb Haemost ; 10(7): 1355-62, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22537188

ABSTRACT

BACKGROUND: The procoagulant properties of microparticles (MPs) are due to the of the presence of phosphatidylserine (PS) and tissue factor (TF) on their surface. The latter has been demonstrated especially on MPs derived from monocytes. OBJECTIVES: To investigate the relative contribution of TF and factor (F)XII in initiating coagulation on MPs derived from monocytes, platelets and erythrocytes. METHODS: Microparticles were isolated from calcium ionophore-stimulated platelets, erythrocytes and monocytic THP-1 cells. MPs were quantified, characterized for cell-specific antigens and analyzed for TF, PS exposure and their thrombin-generating potential. RESULTS: The MP number was not proportional to PS exposure and the majority of the MPs exposed PS. TF activity was undetectable on platelet- and erythrocyte-derived MPs (< 1 fM nM(-1) PS), whereas monocyte-derived MPs exposed TF (32 fM nM(-1) PS). Platelet-, erythrocyte- and monocyte-derived MPs, but not purified phospholipids, initiated thrombin generation in normal plasma in the absence of an external trigger (lag time < 11 min). Deficiency or inhibition of FVII had no effect on thrombin generation induced by platelet- and erythrocyte-derived MPs, but interfered with monocyte MP-triggered coagulation. Platelet- and erythrocyte-derived MPs completely failed to induce thrombin generation in FXII-deficient plasma. In contrast, monocyte-derived MPs induced similar thrombin generation in normal vs. FXII-deficient plasma. CONCLUSION: MPs from platelets and erythrocytes not only propagate coagulation by exposing PS but also initiate thrombin generation independently of TF in a FXII-dependent manner. In contrast, monocyte-derived MPs trigger coagulation predominantly via TF.


Subject(s)
Blood Platelets/physiology , Cell-Derived Microparticles , Erythrocytes/physiology , Factor XIIa/physiology , Thrombin/biosynthesis , Flow Cytometry , Humans , Immunophenotyping , Reference Values
14.
J Thromb Haemost ; 9(7): 1359-67, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21481175

ABSTRACT

BACKGROUND: Particulate matter (PM) is a key component of ambient air pollution and has been associated with an increased risk of thrombotic events and mortality. The underlying mechanisms remain unclear. OBJECTIVES: To study the mechanisms of PM-driven procoagulant activity in human plasma and to investigate mainly, the coagulation driven by ultrafine particles (UFPs; < 0.1 µm) in genetically modified mice. METHODS: Thrombin generation in response to PM of different sizes was assessed in normal human platelet-poor plasma, as well as in plasmas deficient in the intrinsic pathway proteases factors XII (FXII) or XI (FXI). In addition, UFPs were intratracheally instilled in wild-type (WT) and FXII-deficient (FXII(-/-) ) mice and plasma thrombin generation was analyzed in plasma from treated mice at 4 and 20 h post-exposure. RESULTS: In normal human plasma, thrombin generation was enhanced in the presence of PM, whereas PM-driven thrombin formation was completely abolished in FXII- and FXI-deficient plasma. UFPs induced a transient increase in tissue factor (TF)-driven thrombin formation at 4 h post-instillation in WT mice compared with saline instillation. Intratracheal instillation of UFPs resulted in a procoagulant response in WT mice plasma at 20 h, whereas it was entirely suppressed in FXII(-/-) mice. CONCLUSIONS: Overall, the data suggest that PM promotes its early procoagulant actions mostly through the TF-driven extrinsic pathway of coagulation, whereas PM-driven long lasting thrombogenic effects are predominantly mediated via formation of activated FXII. Hence, FXII-driven thrombin formation may be relevant to an enhanced thrombotic susceptibility upon chronic exposure to PM in humans.


Subject(s)
Blood Coagulation , Factor XII/physiology , Particulate Matter/adverse effects , Animals , Enzyme Activation , Factor XI , Factor XII/genetics , Factor XII/metabolism , Mice , Mice, Mutant Strains , Particle Size , Thrombin/biosynthesis , Thrombosis/etiology
15.
J Thromb Haemost ; 9(5): 953-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21352469

ABSTRACT

BACKGROUND: Aortic stenosis patients often show bleeding complications. Previously, a prolonged platelet function analyzer (PFA-100) closure time was observed with plasma of severe aortic stenosis patients. To elucidate a possible role of circulating preactivated von Willebrand factor (VWF), we determined the level of VWF in its active, platelet-binding conformation in plasma of patients with aortic stenosis. PATIENTS/METHODS: Sixty-two aortic stenosis patients were included in this study. VWF and related parameters were measured, and the results were related to severity of aortic stenosis. RESULTS: VWF activation factor, indicating the proportion of circulating VWF able to bind to platelets, correlated negatively with peak transvalvular gradient and PFA-100 closure time. No correlation was observed between ADAMTS13 activity and peak transvalvular gradient or PFA-100 closure time. Both VWF antigen and VWF propeptide levels were significantly higher in patients with mild and moderate aortic stenosis, but not in those with severe stenosis. CONCLUSIONS: Our data demonstrate that the aortic pressure gradient is inversely associated with VWF activation factor, but not with VWF antigen or VWF multimerization in patients with aortic stenosis. These findings might have implications for the bleeding observed in patients with aortic stenosis.


Subject(s)
Aortic Valve Stenosis/blood , Aortic Valve Stenosis/pathology , von Willebrand Factor/biosynthesis , ADAM Proteins/blood , ADAMTS13 Protein , Aged , Female , Hemorrhage , Hemostasis , Humans , Male , Middle Aged , Peptides/chemistry , Platelet Function Tests , Shear Strength , Stress, Mechanical , Surveys and Questionnaires
16.
Neuroimage ; 49(4): 2907-14, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-19958838

ABSTRACT

Intrinsic coagulation factor XII deficient (FXII(-/-)) mice are protected from ischemic stroke. To elucidate underlying mechanisms we investigated the early ischemic period in vivo by multimodal magnetic resonance imaging (MRI) at 17.6 Tesla. Cerebral ischemia was induced by either transient (60 min) or permanent occlusion of the middle cerebral artery (t/pMCAO). 10 FXII(-/-) mice underwent t- , 10 FXII(-/-) mice p- and 10 Wildtype (Wt) mice tMCAO. Cerebral blood flow (CBF), diffusion-weighted-imaging (DWI) and T2-relaxometry were measured at 2 h and 24 h after MCAO. Outcome measures were evaluated after motion correction and normalization to atlas space. 2 h after tMCAO CBF reduction was similar in FXII(-/-) and Wt mice extending over cortical (CBF (ml/100 g/min) 33.6+/-6.9 vs. 35.3+/-4.6, p=0.42) and subcortical regions (25.7+/-4.5 vs. 31.6+/-4.0, p=0.17). At 24 h, recovery of cortical CBF by +36% was observed only in tMCAO FXII(-/-) mice contrasting a further decrease of -30% in Wt mice after tMCAO (p=0.02, F((1,18))=6.24). In FXII(-/-) mice in which patency of the MCA was not restored (pMCAO) a further decrease of -75% was observed. Cortical reperfusion in tMCAO FXII(-/-) mice was related to a lower risk of infarction of 59% vs. 93% in Wt mice (p=0.04). Subcortical CBF was similarly decreased in both tMCAO groups (Wt and FXII(-/-)) relating to a similar risk of infarction of 89% (Wt) vs. 99% (FXII(-/-), p=0.17). Deficiency of FXII allows neocortical reperfusion after tMCAO and rescues brain tissue by this mechanism. This study supports the concept of FXII as a promising new target for stroke prevention and therapy.


Subject(s)
Brain Ischemia/diagnosis , Brain Ischemia/therapy , Brain/pathology , Factor XII Deficiency/therapy , Reperfusion/methods , Stroke/diagnosis , Stroke/therapy , Animals , Brain Ischemia/complications , Disease Models, Animal , Factor XII Deficiency/complications , Factor XII Deficiency/diagnosis , Humans , Mice , Stroke/etiology , Treatment Outcome
17.
J Thromb Haemost ; 7 Suppl 1: 79-83, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19630774

ABSTRACT

The blood coagulation system forms fibrin to limit blood loss from sites of injury, but also contributes to occlusive diseases such as deep vein thrombosis, myocardial infarction, and stroke. In the current model of a coagulation balance, normal hemostasis and thrombosis represent two sides of the same coin; however, data from coagulation factor XI-deficient animal models have challenged this dogma. Gene targeting of factor XI, a serine protease of the intrinsic pathway of coagulation, severely impairs arterial thrombus formation but is not associated with excessive bleeding. Mechanistically, factor XI may be activated by factor XII following contact activation or by thrombin in a feedback activation loop. This review focuses on the role of factor XI, and its deficiency states as novel target for prevention of thrombosis with low bleeding risk in animal models.


Subject(s)
Disease Models, Animal , Factor XI Deficiency , Animals , Factor XI Deficiency/genetics , Hemorrhage/etiology , Hemostasis , Thrombosis/etiology , Thrombosis/prevention & control
18.
J Thromb Haemost ; 6(5): 856-64, 2008 May.
Article in English | MEDLINE | ID: mdl-18284601

ABSTRACT

OBJECTIVES: Platelet hyperreactivity contributes to adverse vascular events in diabetes mellitus. It is unclear whether platelet hyperreactivity is due to impaired insulin effects directly on platelets and /or originates from endothelial cells. Here, acute effects of insulin on platelet activation and platelet-endothelial cell interactions were analyzed. METHODS AND RESULTS: Washed human platelets were treated with insulin alone or in combinations with thrombin, collagen and ADP. Insulin signaling was analyzed by intracellular phosphorylation markers of platelet activation (ERK, p38 MAPK, PKB) or inhibition (VASP), platelet aggregation, intracellular Ca(2+) levels, and platelet adhesion to collagen coated surfaces and endothelial cells under flow. Insulin up to 100 nm for 5 min did not change phosphorylation status of VASP, p38, ERK or PKB in platelets. Integrin alpha(IIb)beta(3) activation, P-selectin expression, aggregation, and platelet adhesion to collagen coated surfaces and endothelial cells under flow were not altered by insulin. An insulin receptor was detected on endothelial cells but not on human platelets. Insulin treatment decreased platelet adhesion to endothelial cells through insulin stimulation of endothelial NO production and NOS inhibition interfered with this process. CONCLUSIONS: Insulin exerts no direct acute effects on platelet function but inhibits platelet-endothelial interaction by insulin stimulation of endothelial NO production.


Subject(s)
Blood Platelets/physiology , Endothelial Cells/physiology , Insulin/pharmacology , Platelet Adhesiveness/drug effects , Endothelial Cells/chemistry , Humans , Nitric Oxide/biosynthesis , Phosphorylation , Platelet Activation , Receptor, Insulin/analysis , Signal Transduction
19.
Br J Pharmacol ; 153(5): 886-93, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17891166

ABSTRACT

BACKGROUND AND PURPOSE: Diabetes-associated vascular dysfunction contributes to increased cardiovascular risk. We investigated whether the phosphodiesterase-5 inhibitor sildenafil would improve vascular function in diabetic rats. EXPERIMENTAL APPROACH: Male Wistar rats were injected with streptozotocin (50 mg kg(-1), i.v.) to induce insulin-deficient diabetes. Direct effects of sildenafil as well as modification of endothelium-dependent and -independent vasorelaxation were investigated in vitro. The effects of acute and chronic (2 week) treatment in vivo of sildenafil on vascular function were also characterized in isolated aortic segments in organ bath chambers 4 weeks after diabetes induction. KEY RESULTS: Sildenafil induced a concentration-dependent vasorelaxation, which was attenuated by the nitric oxide (NO) synthase inhibitor, N(G)-nitro-L-arginine. Acetylcholine-induced endothelium-dependent as well as endothelium-independent relaxation induced by the NO donor, DEA-NONOate, was significantly reduced in aortae from diabetic rats. Incubation with sildenafil in vitro normalized both endothelium-dependent and -independent relaxation in aortae from diabetic rats. Acute as well as chronic in vivo treatment with sildenafil resulted in enhanced endothelium-dependent and -independent vasorelaxation. Superoxide formation was increased in diabetes, associated with enhanced membrane expression of the NAD(P)H oxidase subunit gp91(phox) and Rac, which were both reduced by chronic treatment with sildenafil. CONCLUSIONS AND IMPLICATIONS: We demonstrate that sildenafil treatment rapidly and chronically improves vascular relaxation in diabetic rats. Treatment with sildenafil might provide a similarly beneficial effect in diabetic patients.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Phosphodiesterase Inhibitors/pharmacology , Piperazines/pharmacology , Sulfones/pharmacology , Vasodilation/drug effects , Acetylcholine/pharmacology , Animals , Aorta, Thoracic/metabolism , Diabetes Mellitus, Experimental/physiopathology , Dose-Response Relationship, Drug , Drug Administration Schedule , Endothelium, Vascular/drug effects , Male , Nitric Oxide Synthase/metabolism , Oxidative Stress/drug effects , Phosphodiesterase Inhibitors/administration & dosage , Piperazines/administration & dosage , Purines/administration & dosage , Purines/pharmacology , Rats , Rats, Wistar , Sildenafil Citrate , Streptozocin , Sulfones/administration & dosage , Superoxides/metabolism
20.
J Thromb Haemost ; 5(6): 1106-12, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17388803

ABSTRACT

The classic intrinsic pathway of coagulation is triggered by contact activation of the plasma protease factor (F)XII, followed by sequential proteolytic activation of FX1 and FIX. While a key mechanism for initiating coagulation in some clinically useful in vitro assays, the absence of abnormal bleeding associated with congenital FXII deficiency indicates that the intrinsic pathway is not important for normal blood coagulation in vivo. However, recent work with mice lacking FXII or FXI suggest that these proteases make important contributions to formation of pathologic intravascular thrombi. In models of arterial injury, FXII or FXI null mice are protected from formation of platelet rich occlusive thrombi to a degree similar to that seen in FIX deficient mice (a model for the severe bleeding disorder hemophilia B) or to wild type mice treated with high dose heparin. FXII or FXI deficiency does not appear to prevent the initiation of thrombus formation in these models, but instead causes significant thrombus instability that prevents occlusion of the vessel. These findings raise the possibility that a pathway similar or identical to the intrinsic pathway may operate in vivo under some circumstances. Furthermore, the disproportionate importance of FXII and FXI to occlusive thrombus formation compared to normal hemostasis makes these proteases attractive candidates for therapeutic inhibitors to treat or prevent thromboembolic disorders.


Subject(s)
Blood Coagulation/drug effects , Blood Coagulation/physiology , Thromboembolism/blood , Thromboembolism/drug therapy , Animals , Anticoagulants/therapeutic use , Disease Models, Animal , Factor XI/physiology , Factor XII/physiology , Humans , Mice , Models, Cardiovascular , Thromboplastin/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...