Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
J Am Heart Assoc ; : e034492, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39028040

ABSTRACT

BACKGROUND: Venous thromboembolism is associated with endothelial cell activation that contributes to the inflammation-dependent activation of the coagulation system. Cellular damage is associated with the release of different species of extracellular RNA (eRNA) involved in inflammation and coagulation. TLR3 (toll-like receptor 3), which recognizes (viral) single-stranded or double-stranded RNAs and self-RNA fragments, might be the receptor of these species of eRNA during venous thromboembolism. Here, we investigate how the TLR3/eRNA axis contributes to venous thromboembolism. METHODS AND RESULTS: Thrombus formation and size in wild-type and TLR3 deficient (-/-) mice were monitored by ultrasonography after venous thrombosis induction using the ferric chloride and stasis models. Mice were treated with RNase I, with polyinosinic-polycytidylic acid, a TLR3 agonist, or with RNA extracted from murine endothelial cells. Gene expression and signaling pathway activation were analyzed in HEK293T cells overexpressing TLR3 in response to eRNA or in human umbilical vein endothelial cells transfected with a small interference RNA against TLR3. Plasma clot formation on treated human umbilical vein endothelial cells was analyzed. Thrombosis exacerbated eRNA release in vivo and increased eRNA content within the thrombus. RNase I treatment reduced thrombus size compared with vehicle-treated mice (P<0.05). Polyinosinic-polycytidylic acid and eRNA treatments increased thrombus size in wild-type mice (P<0.01 and P<0.05), but not in TLR3-/- mice, by reinforcing neutrophil recruitment (P<0.05). Mechanistically, TLR3 activation in endothelial cells promotes CXCL5 (C-X-C motif chemokine 5) secretion (P<0.001) and NFκB (nuclear factor kappa-light-chain-enhancer of activated B cells) activation (P<0.05). Finally, eRNA triggered plasma clot formation in vitro (P<0.01). CONCLUSIONS: We show that eRNA and TLR3 activation enhance venous thromboembolism through neutrophil recruitment possibly through secretion of CXCL5, a potent neutrophil chemoattractant.

3.
J Vis Exp ; (134)2018 04 13.
Article in English | MEDLINE | ID: mdl-29708541

ABSTRACT

Venous thrombosis is a common condition affecting 1 - 2% of the population, with an annual incidence of 1 in 500. Venous thrombosis can lead to death through pulmonary embolism or results in the post-thrombotic syndrome, characterized by chronic leg pain, swelling, and ulceration, or in chronic pulmonary hypertension resulting in significant chronic respiratory compromise. This is the most common cardiovascular disease after myocardial infarction and ischemic stroke and is a clinical challenge for all medical disciplines, as it can complicate the course of other disorders such as cancer, systemic disease, surgery, and major trauma. Experimental models are necessary to study these mechanisms. The stasis model induces consistent thrombus size and a quantifiable amount of thrombus. However, it is necessary to systematically ligate side branches of the inferior vena cava to avoid variability in thrombus sizes and any erroneous data interpretation. We have developed a non-invasive technique to measure thrombus size using ultrasonography. Using this technique, we can assess thrombus development and resolution over time in the same animal. This approach limits the number of mice required for quantification of venous thrombosis consistent with the principle of replacement, reduction, and refinement of animals in research. We have demonstrated that thrombus weight and histological analysis of thrombus size correlate with measurement obtained with ultrasonography. Therefore, the current study describes how to induce deep vein thrombosis in mice using the inferior vena cava stasis model and how to monitor it using high frequency ultrasound.


Subject(s)
Ultrasonography/methods , Venous Thrombosis/etiology , Animals , Disease Models, Animal , Male , Mice , Venous Thrombosis/diagnostic imaging , Venous Thrombosis/pathology
4.
Arterioscler Thromb Vasc Biol ; 37(7): 1315-1322, 2017 07.
Article in English | MEDLINE | ID: mdl-28450294

ABSTRACT

OBJECTIVE: Coagulation and inflammation are inter-related. Gas6 (growth arrest-specific 6) promotes venous thrombosis and participates to inflammation through endothelial-innate immune cell interactions. Innate immune cells can provide the initiating stimulus for venous thrombus development. We hypothesize that Gas6 promotes monocyte recruitment during venous thrombosis. APPROACH AND RESULTS: Deep venous thrombosis was induced in wild-type and Gas6-deficient (-/-) mice using 5% FeCl3 and flow reduction in the inferior vena cava. Total monocyte depletion was achieved by injection of clodronate before deep venous thrombosis. Inflammatory monocytes were depleted using an anti-C-C chemokine receptor type 2 (CCR2) antibody. Similarly, injection of an anti-chemokine ligand 2 (CCL2) antibody induced CCL2 depletion. Flow cytometry and immunofluorescence were used to characterize the monocytes recruited to the thrombus. In vivo, absence of Gas6 was associated with a reduction of monocyte recruitment in both deep venous thrombosis models. Global monocyte depletion by clodronate leads to smaller thrombi in wild-type mice. Compared with wild type, the thrombi from Gas6-/- mice contain less inflammatory (CCR2hiCX3CR1lo) monocytes, consistent with a Gas6-dependent recruitment of this monocyte subset. Correspondingly, selective depletion of CCR2hiCX3CR1lo monocytes reduced the formation of venous thrombi in wild-type mice demonstrating a predominant role of the inflammatory monocytes in thrombosis. In vitro, the expression of both CCR2 and CCL2 were Gas6 dependent in monocytes and endothelial cells, respectively, impacting monocyte migration. Moreover, Gas6-dependent CCL2 expression and monocyte migration were mediated via JNK (c-Jun N-terminal kinase). CONCLUSIONS: This study demonstrates that Gas6 specifically promotes the recruitment of inflammatory CCR2hiCX3CR1lo monocytes through the regulation of both CCR2 and CCL2 during deep venous thrombosis.


Subject(s)
Chemotaxis, Leukocyte , Inflammation/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Monocytes/metabolism , Receptors, CCR2/metabolism , Receptors, Chemokine/metabolism , Vena Cava, Inferior/metabolism , Venous Thrombosis/metabolism , Animals , CX3C Chemokine Receptor 1 , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemotaxis, Leukocyte/drug effects , Clodronic Acid/pharmacology , Disease Models, Animal , Endothelial Cells/metabolism , Genetic Predisposition to Disease , Inflammation/genetics , Inflammation/pathology , Inflammation/prevention & control , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Monocytes/drug effects , Paracrine Communication , Phenotype , Receptors, CCR2/genetics , Signal Transduction , Vena Cava, Inferior/drug effects , Vena Cava, Inferior/pathology , Venous Thrombosis/genetics , Venous Thrombosis/pathology , Venous Thrombosis/prevention & control
5.
Blood ; 127(6): 769-77, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26585956

ABSTRACT

Venous thromboembolism is a common complication of cancer. Based on recent evidence that (1) growth arrest-specific 6 (Gas6) regulates the expression of tissue factor during venous thrombosis, and (2) cancer promotes a procoagulant milieu, we hypothesize that Gas6 may be involved in cancer-induced coagulopathy. Venous thrombi were induced in both wild-type (WT) and Gas6-deficient ((-/-)) mice with cancer. WT mice with cancer developed larger thrombi than their healthy counterparts; these larger thrombi induced by cancer were not seen in Gas6(-/-) mice. Whole genome microarray analysis of differential gene expression in WT and Gas6(-/-) endothelial cells exposed to M27 murine lung carcinoma cells reveal that Gas6 increases prostaglandin E synthase (Ptges) expression in endothelial cells. This was confirmed using real-time polymerase chain reaction and immunofluorescence staining. Culture of WT endothelial cells with M27 increases the secretion of prostaglandin E2 (PGE2), the enzymatic product of Ptges, in WT but not in Gas6(-/-) endothelial cells. In WT endothelial cells, Ptges expression was regulated through extracellular signal-regulated kinase 1/2 phosphorylation (ERK1/2). In vitro, PGE2 activates platelets after binding to its receptor, EP3. In vivo, EP3 receptor antagonism reversed the effect of cancer-induced thrombosis in WT mice. These results show that Gas6, through upregulation of PGE2, contributes to cancer-induced venous thrombosis.


Subject(s)
Intercellular Signaling Peptides and Proteins/physiology , Intramolecular Oxidoreductases/genetics , Neoplasms/complications , Venous Thrombosis/etiology , Animals , Cells, Cultured , Dinoprostone/adverse effects , Dinoprostone/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins/genetics , Intramolecular Oxidoreductases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/genetics , Neoplasms/pathology , Prostaglandin-E Synthases , Up-Regulation , Venous Thrombosis/genetics , Venous Thrombosis/pathology
6.
Acta Biomater ; 9(11): 9194-200, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23796406

ABSTRACT

Most fatalities from trauma occur due to severe blood loss. There is a need for improved hemostatic biomaterials that can address this problem. The aim of this study was to determine the in vivo efficacy of nanofibrous microspheres (NFM) loaded with hemostatic peptides, specifically ideal amphipathic peptides (IAP) that have been demonstrated to possess both procoagulant and antifibrinolyic activities. We demonstrate that IAP can be coupled to NFM (IAP-NFM) to form matrices that exhibit substantial hemostatic activity. IAP-NFM matrices were compared to a commercial zeolitic hemostatic biomaterial (QuikClot) and have superior efficacy in reducing bleeding in vivo. In both a murine tail transection and a murine hepatic injury model, bleeding times were significantly reduced (P<0.05) with the use of IAP-NFM as compared with equal masses of either QuikClot or NFM alone, or no treatment. Importantly, histological examination revealed no tissue injury when IAP-NFM or NFM were applied to hepatic lacerations. In contrast, QuikClot caused widespread hepatocyte degeneration and necrosis, with higher average injury zone thickness as determined by semiquantitative analysis. In summary, NFM was able to maintain the pro-coagulant properties of IAP in our preclinical model, caused no observable tissue damage at the site of application and had better performance than QuikClot controls.


Subject(s)
Calcium Phosphates/chemistry , Hemostasis/drug effects , Hemostatics/pharmacology , Microspheres , Nanofibers/chemistry , Peptides/pharmacology , Surface-Active Agents/pharmacology , Adsorption , Animals , Factor X/metabolism , Liver/drug effects , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Tail
7.
Blood ; 121(4): 692-9, 2013 Jan 24.
Article in English | MEDLINE | ID: mdl-23149844

ABSTRACT

Gas6 (growth-arrest specific gene 6) plays a role in thrombus stabilization. Gas6 null (-/-) mice are protected from lethal venous and arterial thromboembolism through platelet signaling defects induced only by 5 µM ADP and 10 µM of the thromboxane analog, U46619. This subtle platelet defect, despite a dramatic clinical phenotype, raises the possibility that Gas6 from a source other than platelets contributes to thrombus formation. Thus, we hypothesize that Gas6 derived from the vascular wall plays a role in venous thrombus formation. Bone marrow transplantation and platelet depletion/reconstitution experiments generating mice with selective ablations of Gas6 from either the hematopoietic or nonhematopoietic compartments demonstrate an approximately equal contribution by Gas6 from both compartments to thrombus formation. Tissue factor expression was significantly reduced in the vascular wall of Gas6(-/-) mice compared with WT. In vitro, thrombin-induced tissue factor expression was reduced in Gas6(-/-) endothelial cells compared with wild-type endothelium. Taken together, these results demonstrate that vascular Gas6 contributes to thrombus formation in vivo and can be explained by the ability of Gas6 to promote tissue factor expression and activity. These findings support the notion that vascular wall-derived Gas6 may play a pathophysiologic role in venous thromboembolism.


Subject(s)
Blood Vessels/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Thromboplastin/genetics , Thrombosis/genetics , Animals , Blood Platelets/metabolism , Blood Vessels/pathology , Endothelial Cells/metabolism , Gene Expression Regulation , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Knockout , Thromboplastin/metabolism , Thrombosis/metabolism
8.
Circ Arrhythm Electrophysiol ; 5(6): 1168-75, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23095228

ABSTRACT

BACKGROUND: The most important complication of atrial fibrillation (AF) is thromboembolic stroke. Although AF-related remodeling is considered important in atrial thrombogenesis, its role never has been directly tested. This study assessed effects of AF-related remodeling on the atrial thrombogenic milieu by using radiofrequency ablation (RFA) to create a quantifiable prothrombotic nidus. METHODS AND RESULTS: We studied normal control dogs (control, n=16) and 3 canine AF-models: (1) atrial tachycardia remodeling (ATR; n=16) induced by atrial tachypacing (400 bpm for 1 week, with atrioventricular block and ventricular pacing at 80 bpm); (2) congestive heart failure (CHF; n=14) attributable to ventricular tachypacing (240 bpm for 2 weeks); and (3) chronic AF (CAF; n=8) induced by atrial tachypacing (35±3 days) without atrioventricular block. CAF dogs had AF for 13±1 days until euthanization. After remodeling was established, RFA lesions were created in both atria. Half the ATR and CHF dogs were subjected to atrial tachypacing during 7-day post-RFA follow-up. Electrophysiological and echocardiographic studies were performed before RFA and 7 days after RFA, and then hearts were removed and atrial thrombi were quantified by histomorphometry. Burst-pacing-induced AF duration was significantly greater in ATR, CHF, and CAF groups versus control group. The atrial effective refractory period shortened in ATR and CAF groups. Left atrial diameter was significantly larger with CHF, but not with ATR. Neither total thrombus volume nor thrombus volume per lesion differed significantly among groups. Table.Properties of Ablation Lesions and Atrial Thrombi Experimental GroupControl (n=16)ATR (n=16)CHF (n=14)CAF (n=8)N of ablation lesions per dog6.9±0.36.6±0.27.2±0.26.9±0.4Ablation lesion area, mm(2)53.1±3.558.3±4.857.7±4.944.3±3.7Ablation lesion depth, mm5.2±0.25.1±0.35.3±0.25.2±0.2Ablation lesion volume, mm(3)205.2±17.8211.6±17.6231.5±29.0176.8±22.2N of thrombi per dog5.4±0.44.7±0.35.6±0.46.5±0.4Presence of thrombus, %80±572±577±695±3Mean thrombus volume in both atria, mm(3)20.8±3.414.9±2.212.2±2.622.5±5.6Mean thrombus volume in left atria, mm(3)8.2±1.54.0±0.95.5±1.68.1±3.3Mean thrombus volume in right atria, mm(3)30.1±5.422.7±4.317.9±4.132.8±8.3Total thrombus volume in both atria, mm(3)140.5±21.399.7±16.886.1±17.5131.1±22.7Total thrombus volume in left atria, mm(3)22.8±5.311.8±3.317.0±3.723.3±6.4Total thrombus volume in right atria, mm(3)117.7±21.587.8±17.269.1±16.1107.8±23.3Thrombus volume normalized to ablation lesion area in both atria, mm(3)/mm(2)0.5±0.10.4±0.11.5±1.10.8±0.3Thrombus volume normalized to ablation lesion volume in both atria0.2±0.10.1±0.00.5±0.40.3±0.1 ATR indicates atrial tachycardia remodeling; CAF, chronic atrial fibrillation; and CHF, congestive heart failure. There were no statistically significant differences for any groups vs control group for any of these variables studied. CONCLUSIONS: None of the AF substrates tested, including sustained atrial tachycardia/AF itself, enhanced post-RFA atrial thrombus formation. Indices of electrical and structural remodeling did not predict post-RFA thrombogenic potential. Contrary to widely held but previously untested notions, we were unable to demonstrate prothrombotic effects of AF-related remodeling.


Subject(s)
Atrial Fibrillation/complications , Atrial Fibrillation/physiopathology , Thrombosis/etiology , Thrombosis/physiopathology , Ventricular Remodeling/physiology , Animals , Antithrombin III , Atrial Fibrillation/blood , C-Reactive Protein/metabolism , Catheter Ablation , Chronic Disease , Disease Models, Animal , Dogs , Electrocardiography , Heart Failure/blood , Heart Failure/physiopathology , Peptide Hydrolases/blood , Thrombosis/blood
9.
Adv Nutr ; 3(2): 196-203, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22516727

ABSTRACT

Gas6 (growth arrest-specific 6) belongs structurally to the family of plasma vitamin K-dependent proteins. Gas6 has a high structural homology with the natural anticoagulant protein S, sharing the same modular composition. Interestingly, despite the presence of a γ-carboxyglutamic acid domain in its structure, no role in the coagulation cascade has been identified for gas6. Gas6 has been shown to be involved in vascular homeostasis and more precisely is involved in proliferation, apoptosis, efferocytosis, leukocyte migration, and sequestration and platelet aggregation. It is also involved in the activation of different cell types, from platelets to endothelial and vascular smooth muscle cells. Thus, it has been shown to play a role in several pathophysiological processes such as atherosclerosis, cancer, and thrombosis. Interestingly, studies using gas6 null mice highlighted that gas6 may represent a novel potential target for anticoagulant therapy, because these animals are protected from lethal venous thromboembolism without excessive bleeding. However, the mechanism in thrombus occurrence remains to be further explored. In the present review, we will focus on the role of gas6 in innate immunity, atherosclerosis, thrombosis, and cancer-related events.


Subject(s)
Hemostasis/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Animals , Arteries/metabolism , Atherosclerosis/metabolism , Humans , Mice , Neoplasms/metabolism , Thrombosis/metabolism , Veins/metabolism
10.
J Trauma Acute Care Surg ; 72(1): 136-42, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22027883

ABSTRACT

BACKGROUND: The leading causes of death for trauma patients in civilian and combat settings are traumatic brain injury and uncontrolled hemorrhage, respectively. This study examines the hemostatic activity of an ideal amphipathic peptide (IAP) attached to a biocompatible surface. METHODS: Procoagulant properties of IAP attached to a surface were first tested, in vitro, using factor Xa and thrombin generation assays and thromboelastography. Rabbits and swine were used for in vivo studies. Injuries were performed using scalpel blade 11, and free bleeding was allowed for five seconds. While bleeding, IAP coupled to a hydrogel or QuikClot were applied to the wound and the time was recorded until bleeding stopped. RESULTS: Results show that when IAP is attached to a surface, both factor IXa and factor Xa activities are promoted. Thromboelastography shows that surface-attached IAP results in earlier onset and stronger clot formation. In rabbits, the incorporation of IAP onto a biocompatible hydrogel reduces bleeding times by 40% (p < 0.03). In pigs, bleeding times are reduced 30% to 50% (p < 0.02) by surface-coupled IAP. Finally, using a rabbit liver laceration model, the properties of surface-coupled IAP are less damaging when compared with QuikClot, a currently used material in external hemorrhagic injuries. CONCLUSIONS: This study provides a relevant proof of concept for the development of IAP coupled to a biocompatible surface as a hemostatic agent, that is potentially safer than the commercially available QuikClot.


Subject(s)
Bandages, Hydrocolloid , Brain Hemorrhage, Traumatic/drug therapy , Hemostatics/therapeutic use , Peptides/therapeutic use , Animals , Ear, External/injuries , Factor Xa/analysis , Femoral Artery/injuries , Hemostatics/administration & dosage , Liver/injuries , Male , Peptides/administration & dosage , Rabbits , Swine , Thrombelastography , Thrombin/analysis
11.
Clin Appl Thromb Hemost ; 18(1): 27-34, 2012.
Article in English | MEDLINE | ID: mdl-21873363

ABSTRACT

In this study, we validated a thromboelastography (TEG) method to evaluate the hemostatic effects of 3 peptides. The first peptide is an ideal amphipathic peptide composed of 22 leucine and lysine in a ratio of 2:1. At a very low concentration, the peptide had a procoagulant effect shown by decreases in reaction time (R) and coagulation time (K) but was impaired by a decrease in maximum amplitude (MA). At higher concentrations, the peptide had an anticoagulant effect. The α angle was minimally affected by the peptide. The second peptide is melittin derived from bee venom. Melittin showed procoagulant effects reflected by a decrease in clotting time but led to lower MA. The third peptide derived from fibrinogen γ chain promoted hemostasis only at an optimal concentration and became anticoagulant at a higher concentration. The hemostatic mechanisms of each peptide were discussed. Our study would facilitate further development of peptides for either hemorrhage control or thrombosis treatment.


Subject(s)
Blood Coagulation/drug effects , Fibrinogen/pharmacology , Hemostatics/pharmacology , Melitten/pharmacology , Thrombelastography , Dose-Response Relationship, Drug , Fibrinogen/chemistry , Hemostatics/chemistry , Humans , Melitten/chemistry
12.
Blood Coagul Fibrinolysis ; 22(7): 622-3, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21760480

ABSTRACT

Heyde's syndrome is characterized by iron deficiency anemia due to gastrointestinal bleeding and calcific aortic stenosis. Patients with this syndrome have a bleeding diathesis due to a loss of the largest multimers of von Willebrand factor (vWF). Here we present a case of Heyde's syndrome diagnosed with abnormal closure times and normal vWF Ristocetin cofactor activity (vWF:Rco). In this case, a 79-year-old man with known aortic stenosis and recurrent gastrointestinal bleeding was cured of a life-threatening hemorrhage after the replacement of his stenotic aortic valve. Factor VIII activity (1.53  IU/ml), vWF antigen (1.26  IU/ml), vWF:Rco (1.11  IU/ ml), and the ratio of vWF antigen/vWF:Rco (0.88) were all within normal limits. Instead, to prove a defect in platelet aggregation, closure times measured with collagen/ADP and collagen/epinephrine were abnormal (>300  s). Postoperatively, these closure times normalized. What is unique about our current report is that we measured both vWF:Rco and closure times, the two readily available assays in most coagulation laboratories. vWF:Rco is a standard assay for measuring platelet activity but may miss defects in platelet aggregation that are only seen under high shear stress. As the closure times can detect such defects, it is perhaps more representative than traditional assays, and in situations such as our case, closure times may be the only method by which subtle abnormalities in vWF function could be detected.


Subject(s)
Aortic Valve Stenosis/physiopathology , Aortic Valve/physiopathology , Blood Coagulation Tests/methods , Gastrointestinal Hemorrhage/physiopathology , von Willebrand Diseases/diagnosis , Adenosine Diphosphate/metabolism , Aged , Aortic Valve/pathology , Aortic Valve/surgery , Aortic Valve Stenosis/complications , Aortic Valve Stenosis/pathology , Aortic Valve Stenosis/surgery , Collagen/chemistry , Epinephrine/metabolism , Factor VIII/analysis , Gastrointestinal Hemorrhage/complications , Gastrointestinal Hemorrhage/pathology , Gastrointestinal Hemorrhage/surgery , Humans , Male , Platelet Aggregation , Ristocetin/analysis , von Willebrand Diseases/blood , von Willebrand Diseases/complications , von Willebrand Diseases/pathology , von Willebrand Diseases/physiopathology , von Willebrand Factor/analysis
13.
J Thromb Thrombolysis ; 32(3): 272-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21638224

ABSTRACT

Growth arrest-specific 6 (gas6), a novel vitamin K-dependent protein, has been demonstrated to have a role in thrombus stabilization as gas6 null mice are resistant to lethal venous and arterial thrombosis. However, the association between gas6 and venous thromboembolism has not been elucidated in humans. The present study aims to assess the role of gas6 in human venous thromboembolic (VTE) disease. Using a highly specific ELISA method, we measured plasma levels of gas6 in plasma samples obtained from 279 patients with VTE and 79 healthy volunteers. Medication history, comorbid conditions and VTE characteristics were documented. Mean gas6 levels were higher in patients with VTE as compared to healthy volunteers, being 46 ±11 ng/ml and 35 ±6.4 ng/ml respectively (P < 0.001). Odds ratios (OR) for VTE given elevated (≥90th percentile of healthy volunteers) gas6 levels were estimated in regression models in the whole study population. After adjustment for age, sex, medications and comorbidity, subjects with elevated gas6 had an increased risk of VTE (OR of 16.3 (95% CI 5.8-45.7, P < 0.001) compared to those with lower levels of gas6. This association remains significant even among patients with a comparable age distribution. Among patients with VTE, mean gas levels showed a trend of higher levels in those with more extensive thrombi. There was no correlation between elevated gas6 levels and recurrent VTE. In conclusion, we demonstrate an association between VTE and elevated gas6 levels consistent with in vivo murine models of thrombosis. This constitutes a potential novel mechanism for thrombosis in humans and may aid in the understanding of the pathophysiology of VTE.


Subject(s)
Intercellular Signaling Peptides and Proteins/blood , Venous Thromboembolism/blood , Adult , Aged , Animals , Biomarkers/blood , Cohort Studies , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Mice , Middle Aged , Risk Factors
14.
Biomaterials ; 32(1): 295-305, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20864158

ABSTRACT

Gene therapy for hemophilia B and other hereditary plasma protein deficiencies showed great promise in pre-clinical and early clinical trials. However, safety concerns about in vivo delivery of viral vectors and poor post-transplant survival of ex vivo modified cells remain key hurdles for clinical translation of gene therapy. We here describe a 3D scaffold system based on porous hydroxyapatite-PLGA composites coated with biomineralized collagen 1. When combined with autologous gene-engineered factor IX (hFIX) positive mesenchymal stem cells (MSCs) and implanted in hemophilic mice, these scaffolds supported long-term engraftment and systemic protein delivery by MSCs in vivo. Optimization of the scaffolds at the macro-, micro- and nanoscales provided efficient cell delivery capacity, MSC self-renewal and osteogenesis respectively, concurrent with sustained delivery of hFIX. In conclusion, the use of gene-enhanced MSC-seeded scaffolds may be of practical use for treatment of hemophilia B and other plasma protein deficiencies.


Subject(s)
Genetic Therapy/methods , Hemophilia B/therapy , Mesenchymal Stem Cells/metabolism , Tissue Scaffolds/chemistry , Animals , Calcium Phosphates/pharmacology , Cell Lineage/drug effects , Cell Proliferation/drug effects , Ceramics/pharmacology , Factor IX/genetics , Factor IX/therapeutic use , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/ultrastructure , Mice , Nanoparticles/ultrastructure , Particle Size , Porosity/drug effects
15.
Am J Physiol Heart Circ Physiol ; 300(3): H745-53, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21169404

ABSTRACT

Hyperhomocysteinemia (HHcy) has been shown to induce endothelial dysfunction in part as a result of enhanced oxidative stress. Function and survival of endothelial progenitor cells (EPCs, defined as sca1(+) c-kit(+) flk-1(+) bone marrow-derived cells), which significantly contribute to neovascularization and endothelial regeneration, depend on controlled production of reactive oxygen species (ROS). Mice heterozygous for the gene deletion of methylenetetrahydrofolate reductase (Mthfr(+/-)) have a 1.5- to 2-fold elevation in plasma homocysteine. This mild HHcy significantly reduced the number of circulating EPCs as well as their differentiation. Mthfr deficiency was also associated with increased ROS production and reduced nitric oxide (NO) generation in Mthfr(+/-) EPCs. Treatment of EPCs with sepiapterin, a precursor of tetrahydrobiopterin (BH(4)), a cofactor of endothelial nitric oxide synthase (eNOS), significantly reduced ROS and improved NO production. mRNA and protein expression of eNOS and the relative amount of eNOS dimer compared with monomer were decreased by Mthfr deficiency. Impaired differentiation of EPCs induced by Mthfr deficiency correlated with increased senescence, decreased telomere length, and reduced expression of SIRT1. Addition of sepiapterin maintained cell senescence and SIRT1 expression at levels comparable to the wild type. Taken together, these results demonstrate that Mthfr deficiency impairs EPC formation and increases EPC senescence by eNOS uncoupling and downregulation of SIRT1.


Subject(s)
Cellular Senescence/genetics , Endothelial Cells/enzymology , Nitric Oxide Synthase Type III/metabolism , Sirtuin 1/metabolism , Stem Cells/enzymology , Animals , Cell Differentiation/genetics , Down-Regulation , Female , Homocystinuria/genetics , Hyperhomocysteinemia/drug therapy , Hyperhomocysteinemia/enzymology , Hyperhomocysteinemia/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Mice , Muscle Spasticity/genetics , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Oxidative Stress/genetics , Psychotic Disorders/genetics , Pterins/pharmacology , Reactive Oxygen Species , Telomere/metabolism
16.
Thromb Haemost ; 104(1): 118-21, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20458438

ABSTRACT

Unanticipated elevation of the INR is common in patients receiving warfarin. We performed a prospective cohort study of 107 warfarin-treated patients with INR values of more than 10 who received a single 2.5 mg dose of oral vitamin K. During the first week, one patient experienced major bleeding, and one died. In the first 90 days after enrolment four patients had major bleeding (3.7%, 1.0% to 9.3%), eight patients (7.5%, 3.3% to 14.2%) died and two had objectively confirmed thromboembolism. Based on our low rate of observed major bleeding we conclude that 2.5 mg of oral vitamin K is a reasonable treatment for patients with INR values of more than 10 who are not actively bleeding.


Subject(s)
Anticoagulants/administration & dosage , Venous Thrombosis/drug therapy , Vitamin K/administration & dosage , Warfarin/administration & dosage , Administration, Oral , Aged , Aged, 80 and over , Anticoagulants/adverse effects , Female , Follow-Up Studies , Hemorrhage/etiology , Hemorrhage/prevention & control , Humans , International Normalized Ratio , Male , Middle Aged , Prospective Studies , Venous Thrombosis/blood , Venous Thrombosis/physiopathology , Vitamin K/adverse effects , Warfarin/adverse effects
17.
Ann Intern Med ; 150(5): 293-300, 2009 Mar 03.
Article in English | MEDLINE | ID: mdl-19258557

ABSTRACT

BACKGROUND: Low-dose oral vitamin K decreases the international normalized ratio (INR) in overanticoagulated patients who receive warfarin therapy. Its effects on bleeding events are uncertain. OBJECTIVE: To see whether low-dose oral vitamin K reduces bleeding events over 90 days in patients with warfarin-associated coagulopathy. DESIGN: Multicenter, randomized, placebo-controlled trial. Randomization was computer-generated, and participants were allocated to trial groups by using sequentially numbered study drug containers. Patients, caregivers, and those who assessed outcomes were blinded to treatment assignment. SETTING: 14 anticoagulant therapy clinics in Canada, the United States, and Italy. PATIENTS: Nonbleeding patients with INR values of 4.5 to 10.0. INTERVENTION: Oral vitamin K, 1.25 mg (355 patients randomly assigned; 347 analyzed), or matching placebo (369 patients randomly assigned; 365 analyzed). MEASUREMENTS: Bleeding events (primary outcome), thromboembolism, and death (secondary outcomes). RESULTS: 56 patients (15.8%) in the vitamin K group and 60 patients (16.3%) in the placebo group had at least 1 bleeding complication (absolute difference, -0.5 percentage point [95% CI, -6.1 to 5.1 percentage points]); major bleeding events occurred in 9 patients (2.5%) in the vitamin K group and 4 patients (1.1%) in the placebo group (absolute difference, 1.5 percentage points [CI, -0.8 to 3.7 percentage points]). Thromboembolism occurred in 4 patients (1.1%) in the vitamin K group and 3 patients (0.8%) in the placebo group (absolute difference, 0.3 percentage point [CI, -1.4 to 2.0 percentage points]). Other adverse effects were not assessed. The day after treatment, the INR had decreased by a mean of 1.4 in the placebo group and 2.8 in the vitamin K group (P < 0.001). LIMITATION: Patients who were actively bleeding were not included, and warfarin dosing after enrollment was not mandated or followed. CONCLUSION: Low-dose oral vitamin K did not reduce bleeding in warfarin recipients with INRs of 4.5 to 10.0. FUNDING: Canadian Institutes of Health Research and Italian Ministry of Universities and Research.


Subject(s)
Anticoagulants/adverse effects , Antifibrinolytic Agents/administration & dosage , Hemorrhage/drug therapy , Vitamin K/administration & dosage , Warfarin/adverse effects , Administration, Oral , Age Factors , Aged , Aged, 80 and over , Female , Hemorrhage/chemically induced , Hemorrhage/prevention & control , Humans , International Normalized Ratio , Male , Middle Aged , Placebos , Thromboembolism/prevention & control , Treatment Outcome
18.
J Biomed Mater Res B Appl Biomater ; 89(1): 199-209, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18777584

ABSTRACT

The fabrication of a novel in situ forming hydrogel composed of a multifunctional poly(ethylene glycol) (PEG) N-hydroxysuccinimide ester (NHS) and poly(allylamine hydrochloride) (PAA) was investigated. FTIR confirmed that PAA formed the hydrogel matrix (i.e., the formation of a PAA-like hydrogel). A factorial experiment was conducted to identify the key parameters that controlled gelation time, gel content, and swelling properties. The type of PEG (e.g., 4- and 6-arm) appeared to play a major role in determining all three performance parameters, with the greatest effect on gelation time. Other influencing factors include (a) the PEG concentration, which contributes to the gelation time and gel content; (b) pH of the buffer used for dissolving each polymer, which can affect the gelation time; and (c) PAA molecular weights, which contribute to the gel content and swelling. The concentration of PAA solution had no significant effects on hydrogel formation and properties within the investigated range, presumably due to negligible changes in the crosslinking density of the hydrogels. The PAA buffer pH influenced the gel content as well. Finally, thromboelastography was used to examine the effects of each polymer and their in situ gelation on blood coagulation in vitro. All individual polymers tested reduced clot strength, while the gelation of the polymers enhanced overall procoagulant effects. These results suggest that the biomaterial can be optimized to provide a combination of rapid gelation and swelling properties suitable for hemorrhage control and thus warrant further studies in animal bleeding models.


Subject(s)
Biocompatible Materials , Hemorrhage/therapy , Hydrogels , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Biocompatible Materials/therapeutic use , Blood Coagulation/drug effects , Esters/chemistry , Esters/pharmacology , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , Hydrogels/therapeutic use , Molecular Structure , Polyamines/chemistry , Polyamines/pharmacology , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Succinimides/chemistry , Succinimides/pharmacology , Viscosity
19.
J Biol Chem ; 283(26): 17991-8001, 2008 Jun 27.
Article in English | MEDLINE | ID: mdl-18450759

ABSTRACT

The modification of glutamic acid residues to gamma-carboxyglutamic acid (Gla) is a post-translational modification catalyzed by the vitamin K-dependent enzyme gamma-glutamylcarboxylase. Despite ubiquitous expression of the gamma-carboxylation machinery in mammalian tissues, only 12 Gla-containing proteins have so far been identified in humans. Because bone tissue is the second most abundant source of Gla-containing proteins after the liver, we sought to identify Gla proteins secreted by bone marrow-derived mesenchymal stromal cells (MSCs). We used a proteomics approach to screen the secretome of MSCs with a combination of two-dimensional gel electrophoresis and tandem mass spectrometry. The most abundant Gla-containing protein secreted by MSCs was identified as periostin, a previously unrecognized gamma-carboxylated protein. In silico amino acid sequence analysis of periostin demonstrated the presence of four consensus gamma-carboxylase recognition sites embedded within fasciclin-like protein domains. The carboxylation of periostin was confirmed by immunoprecipitation and purification of the recombinant protein. Carboxylation of periostin could be inhibited by warfarin in MSCs, demonstrating its dependence on the presence of vitamin K. We were able to demonstrate localization of carboxylated periostin to bone nodules formed by MSCs in vitro, suggesting a role in extracellular matrix mineralization. Our data also show that another fasciclin I-like protein, betaig-h3, contains Gla. In conclusion, periostin is a member of a novel vitamin K-dependent gamma-carboxylated protein family characterized by the presence of fasciclin domains. Furthermore, carboxylated periostin is produced by bone-derived cells of mesenchymal lineage and is abundantly found in mineralized bone nodules in vitro.


Subject(s)
Cell Adhesion Molecules/physiology , Gene Expression Regulation , Mesoderm/cytology , Stromal Cells/cytology , Vitamin K/chemistry , Amino Acid Sequence , Animals , Cell Adhesion Molecules/metabolism , Cell Differentiation , Humans , Mice , Mice, Inbred C57BL , Models, Biological , Molecular Sequence Data , Osteoblasts/metabolism , Sequence Homology, Amino Acid , Stromal Cells/metabolism
20.
Biochem J ; 412(3): 545-51, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18336367

ABSTRACT

On the basis of previous evidence that amphipathic helical peptides accelerate Factor IXa activation of Factor X [Blostein, Rigby, Furie, Furie and Gilbert (2000) Biochemistry 39, 12000-12006], the present study was designed to assess the procoagulant activity of an IAP (ideal amphipathic peptide) of Lys(7)Leu(15) composition. The results show that IAP accelerates Factor X activation by Factor IXa in a concentration-dependent manner and accelerates thrombin generation by Factor Xa with a comparable peptide- and substrate-concentration-dependence. A scrambled helical peptide with the same amino acid composition as IAP, but with its amphipathicity abolished, eliminated most of the aforementioned effects. The Gla (gamma-carboxyglutamic acid)-rich domain of Factor X is required for IAP activity, suggesting that this peptide behaves as a phospholipid membrane. This hypothesis was confirmed, using fluorescence spectroscopy, by demonstrating direct binding between IAP and the Gla-rich domain of Factor X. In addition, the catalytic efficiencies of the tenase and prothrombinase enzymatic complexes, containing cofactors Factor VIIIa and Factor Va respectively, are enhanced by IAP. Finally, we show that IAP delays clot lysis in vitro. In summary, these observations demonstrate that IAP not only enhances essential procoagulant reactions required for fibrin generation, but also inhibits fibrinolysis, suggesting a potential role for IAP as a haemostatic agent.


Subject(s)
Blood Coagulation Factors/metabolism , Coagulants/chemistry , Coagulants/pharmacology , Peptides/chemistry , Peptides/pharmacology , Amino Acid Sequence , Blood Coagulation/physiology , Blood Coagulation Factors/chemistry , Factor IXa/metabolism , Factor X/metabolism , Fibrinolysis , Kinetics , Molecular Sequence Data , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL
...