Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Blood Adv ; 8(2): 441-452, 2024 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-37773781

RESUMO

ABSTRACT: Hemophilia B (HB) is caused by an inherited deficiency of plasma coagulation factor IX (FIX). Approximately 60% of pediatric patients with HB possess a severe form of FIX deficiency (<1% FIX activity). Treatment typically requires replacement therapy through the administration of FIX. However, exogenous FIX has a limited functional half-life, and the natural anticoagulant protein S (PS) inhibits activated FIX (FIXa). PS ultimately limits thrombin formation, which limits plasma coagulation. This regulation of FIXa activity by PS led us to test whether inhibiting PS would extend the functional half-life of FIX and thereby prolong FIX-based HB therapy. We assayed clotting times and thrombin generation to measure the efficacy of a PS antibody for increasing FIX activity in commercially obtained plasma and plasma from pediatric patients with HB. We included 11 pediatric patients who lacked additional comorbidities and coagulopathies. In vivo, we assessed thrombus formation in HB mice in the presence of the FIXa ± PS antibody. We found an accelerated rate of clotting in the presence of PS antibody. Similarly, the peak thrombin formed was significantly greater in the presence of the PS antibody, even in plasma from patients with severe HB. Furthermore, HB mice injected with PS antibody and FIX had a 4.5-fold higher accumulation of fibrin at the thrombus induction site compared with mice injected with FIX alone. Our findings imply that a PS antibody would be a valuable adjunct to increase the effectiveness of FIX replacement therapy in pediatric patients who have mild, moderate, and severe HB.


Assuntos
Hemofilia B , Trombose , Humanos , Camundongos , Criança , Animais , Hemofilia B/tratamento farmacológico , Trombina/metabolismo , Fator IX/uso terapêutico , Fator IX/metabolismo , Fator IXa/metabolismo , Anticorpos
2.
Thromb Res ; 228: 163-171, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37331119

RESUMO

INTRODUCTION: Arterial thrombosis is the main underlying mechanism of acute atherothrombosis. Combined antiplatelet and anticoagulant regimens prevent thrombosis but increase bleeding rates. Mast cell-derived heparin proteoglycans have local antithrombotic properties, and their semisynthetic dual AntiPlatelet and AntiCoagulant (APAC) mimetic may provide a new efficacious and safe tool for arterial thrombosis. We investigated the in vivo impact of intravenous APAC (0.3-0.5 mg/kg; doses chosen according to pharmacokinetic studies) in two mouse models of arterial thrombosis and the in vitro actions in mouse platelets and plasma. MATERIALS AND METHODS: Platelet function and coagulation were studied with light transmission aggregometry and clotting times. Carotid arterial thrombosis was induced either by photochemical injury or surgically exposing vascular collagen after infusion of APAC, UFH or vehicle. Time to occlusion, targeting of APAC to the vascular injury site and platelet deposition on these sites were assessed by intra-vital imaging. Tissue factor activity (TF) of the carotid artery and in plasma was captured. RESULTS: APAC inhibited platelet responsiveness to agonist stimulation (collagen and ADP) and prolonged APTT and thrombin time. After photochemical carotid injury, APAC-treatment prolonged times to occlusion in comparison with UFH or vehicle, and decreased TF both in carotid lysates and plasma. Upon binding from circulation to vascular collagen-exposing injury sites, APAC reduced the in situ platelet deposition. CONCLUSIONS: Intravenous APAC targets arterial injury sites to exert local dual antiplatelet and anticoagulant actions and attenuates thrombosis upon carotid injuries in mice. Systemic APAC provides local efficacy, highlighting APAC as a novel antithrombotic to reduce cardiovascular complications.


Assuntos
Trombose das Artérias Carótidas , Trombose , Lesões do Sistema Vascular , Animais , Camundongos , Anticoagulantes/farmacologia , Anticoagulantes/uso terapêutico , Anticoagulantes/química , Tromboplastina , Inibidores da Agregação Plaquetária/farmacologia , Inibidores da Agregação Plaquetária/uso terapêutico , Fibrinolíticos/uso terapêutico , Trombose/etiologia , Trombose das Artérias Carótidas/tratamento farmacológico , Colágeno/farmacologia , Agregação Plaquetária
3.
Mol Ther Nucleic Acids ; 31: 440-451, 2023 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-36817726

RESUMO

Pathological blood clotting, or thrombosis, limits vital blood flow to organs; such deprivation can lead to catastrophic events including myocardial infarction, pulmonary embolism, and ischemic stroke. Prompt restoration of blood flow greatly improves outcomes. We explored whether aptamers could serve as molecular imaging probes to rapidly detect thrombi. An aptamer targeting thrombin, Tog25t, was found to rapidly localize to and visualize pre-existing clots in the femoral and jugular veins of mice using fluorescence imaging and, when circulating, was able to image clots as they form. Since free aptamer is quickly cleared from circulation, contrast is rapidly developed, allowing clot visualization within minutes. Moreover, administration of an antidote oligonucleotide further enhanced contrast development, causing the unbound aptamer to clear within 5min while impacting the clot-bound aptamer more slowly. These findings suggest that aptamers can serve as imaging agents for rapid detection of thrombi in acute care and perioperative settings.

4.
Bioeng Transl Med ; 7(2): e10277, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35600656

RESUMO

Targeted drug delivery for maintaining blood fluidity can reduce the risks associated with systemic anticoagulants that can lead to off-target bleeding. Recently, there has been much interest in targeted delivery of tissue-type plasminogen activator (tPA) for treating thrombotic complications. The work presented here characterizes a fibrin-specific nanogel (FSN) design for targeted delivery of tPA to treat thrombotic complications. Fibrin binding and clot degradation were characterized in vitro, and animal models of thrombosis were used to examine nanogel effects on coagulation parameters. In vitro assays showed tPA-FSNs attach to fibrin in a dose-dependent manner independent of tPA loading. In animal models of thrombosis, including an electrolytic injury to monitor clot properties in real time, and a lipopolysaccharide-induced disseminated intravascular coagulation (DIC) animal model, tPA-FSNs modulated fibrin/fibrinogen and platelet incorporation into clots and at optimized dosing could recover consumptive coagulopathy in DIC. Distribution of unloaded and tPA-loaded FSNs showed potential clearance of tPA-FSNs after 24 h, although unloaded FSNs may be retained at sites of fibrin deposits. Maximum tolerated dose studies showed tPA-FSNs have minimal toxicity up to 20 times the optimized therapeutic dose. Overall, these studies demonstrate the therapeutic efficacy of targeted fibrinolysis for systemic microthrombi and begin to evaluate key translational parameters for tPA-FSN therapeutics, including optimal tPA-FSN dosage in a DIC rodent model and safety of intravenous tPA-FSN therapeutics.

5.
Blood ; 139(21): 3194-3203, 2022 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-35358299

RESUMO

Platelets are critical in hemostasis and a major contributor to arterial thrombosis (AT). (Pre)clinical studies suggest platelets also contribute to venous thrombosis (VT), but the mechanisms are largely unknown. We hypothesized that in VT, platelets use signaling machinery distinct from AT. Here we aimed to characterize the contributions of platelet G protein-coupled (GPCR) and immunoreceptor tyrosine-based activation motif (ITAM) receptor signaling to VT. Wild-type (WT) and transgenic mice were treated with inhibitors to selectively inhibit platelet-signaling pathways: ITAM-CLEC2 (Clec2mKO), glycoprotein VI (JAQ1 antibody), and Bruton's tyrosine kinase (ibrutinib); GPCR-cyclooxygenase 1 (aspirin); and P2Y12 (clopidogrel). VT was induced by inferior vena cava stenosis. Thrombin generation in platelet-rich plasma and whole-blood clot formation were studied ex vivo. Intravital microscopy was used to study platelet-leukocyte interactions after flow restriction. Thrombus weights were reduced in WT mice treated with high-dose aspirin + clopidogrel (dual antiplatelet therapy [DAPT]) but not in mice treated with either inhibitor alone or low-dose DAPT. Similarly, thrombus weights were reduced in mice with impaired ITAM signaling (Clec2mKO + JAQ1; WT + ibrutinib) but not in Clec2mKO or WT + JAQ1 mice. Both aspirin and clopidogrel, but not ibrutinib, protected mice from FeCl3-induced AT. Thrombin generation and clot formation were normal in blood from high-dose DAPT- or ibrutinib-treated mice; however, platelet adhesion and platelet-neutrophil aggregate formation at the vein wall were reduced in mice treated with high-dose DAPT or ibrutinib. In summary, VT initiation requires platelet activation via GPCRs and ITAM receptors. Strong inhibition of either signaling pathway reduces VT in mice.


Assuntos
Trombose , Trombose Venosa , Animais , Aspirina , Plaquetas/metabolismo , Clopidogrel/metabolismo , Clopidogrel/farmacologia , Proteínas de Ligação ao GTP , Motivo de Ativação do Imunorreceptor Baseado em Tirosina , Camundongos , Camundongos Transgênicos , Ativação Plaquetária , Agregação Plaquetária , Inibidores da Agregação Plaquetária/farmacologia , Trombina/metabolismo , Trombose/metabolismo , Trombose Venosa/metabolismo
6.
J Thromb Haemost ; 19(12): 2997-3007, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34431201

RESUMO

BACKGROUND: Venous thrombosis (VT) and pulmonary embolism (PE), collectively venous thromboembolism (VTE), cause high mortality and morbidity. Factor XIII (FXIII) crosslinks fibrin to enhance thrombus stability and consequently may influence PE risk. Elucidating mechanisms contributing to PE is limited by a lack of models that recapitulate human PE characteristics. OBJECTIVE: We aimed to develop a mouse model that permits embolization of red blood cell (RBC)- and fibrin-rich VT and determine the contribution of FXIII to PE risk. METHODS AND RESULTS: In a thrombin-infusion PE model, F13a+/+ , F13a+/- , and F13a-/-  mice had similar incidence of microthrombi in the lungs; however, thrombi were small, with low RBC content (≤7%), unlike human PEs (~70%). To identify a model producing PE consistent with histological characteristics of human PE, we compared mouse femoral vein electrolytic injury, femoral vein FeCl3 injury, and infrarenal vena cava (IVC) stasis models of VT. Electrolytic and FeCl3  models produced small thrombi with few RBCs (5% and 4%, respectively), whereas IVC stasis produced large thrombi with higher RBC content (68%) that was similar to human PEs. After IVC stasis and ligature removal (de-ligation) to permit thrombus embolization, compared to F13a+/+ mice, F13a+/-  and F13a-/-  mice had similar and increased PE incidence, respectively. CONCLUSIONS: Compared to thrombin infusion-, electrolytic injury-, and FeCl3 -based models, IVC stasis produces thrombi that are more histologically similar to human thrombi. IVC stasis followed by de-ligation permits embolization of existing RBC- and fibrin-rich thrombi. Complete FXIII deficiency increases PE incidence, but partial deficiency does not.


Assuntos
Deficiência do Fator XIII , Embolia Pulmonar , Tromboembolia Venosa , Trombose Venosa , Animais , Modelos Animais de Doenças , Fator XIII/genética , Camundongos , Camundongos Knockout
7.
Biomater Sci ; 9(15): 5160-5174, 2021 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-34312627

RESUMO

Lack of long-term patency has hindered the clinical use of small-diameter prosthetic vascular grafts with the majority of these failures due to the development of neointimal hyperplasia. Previous studies by our laboratory revealed that small-diameter expanded polytetrafluoroethylene (ePTFE) grafts coated with antioxidant elastomers are a promising localized therapy to inhibit neointimal hyperplasia. This work is focused on the development of poly(diol-co-citrate-co-ascorbate) (POCA) elastomers with tunable properties for coating ePTFE vascular grafts. A bioactive POCA elastomer (@20 : 20 : 8, [citrate] : [diol] : [ascorbate]) coating was applied on a 1.5 mm diameter ePTFE vascular graft as the most promising therapeutic candidate for reducing neointimal hyperplasia. Surface ascorbate density on the POCA elastomer was increased to 67.5 ± 7.3 ng mg-1 cm-2. The mechanical, antioxidant, biodegradable, and biocompatible properties of POCA demonstrated desirable performance for in vivo use, inhibiting human aortic smooth muscle cell proliferation, while supporting human aortic endothelial cells. POCA elastomer coating number was adjusted by a modified spin-coating method to prepare small-diameter ePTFE vascular grafts similar to natural vessels. A significant reduction in neointimal hyperplasia was observed after implanting POCA-coated ePTFE vascular grafts in a guinea pig aortic interposition bypass graft model. POCA elastomer thus offers a new avenue that shows promise for use in vascular engineering to improve long-term patency rates by coating small-diameter ePTFE vascular grafts.


Assuntos
Elastômeros , Politetrafluoretileno , Animais , Prótese Vascular , Citratos , Ácido Cítrico , Células Endoteliais/patologia , Cobaias , Hiperplasia/prevenção & controle
8.
Blood Adv ; 5(2): 487-495, 2021 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-33496742

RESUMO

Pancreatic cancer patients have a high risk of venous thromboembolism (VTE). Plasminogen activator inhibitor 1 (PAI-1) inhibits plasminogen activators and increases the risk of thrombosis. PAI-1 is expressed by pancreatic tumors and human pancreatic cell lines. However, to date, there are no studies analyzing the association of active PAI-1 and VTE in pancreatic cancer patients. We investigated the association of active PAI-1 in plasma and VTE in pancreatic cancer patients. In addition, we determined if the presence of human pancreatic tumors expressing PAI-1 impairs venous thrombus resolution in mice. Plasma levels of active PAI-1 in patients with pancreatic cancer and mice bearing human tumors were determined by enzyme-linked immunosorbent assay. We measured PAI-1 expression in 5 different human pancreatic cancer cell lines and found that PANC-1 cells expressed the highest level. PANC-1 tumors were grown in nude mice. Venous thrombosis was induced by complete ligation of the inferior vena cava (IVC). Levels of active PAI-1 were independently associated with increased risk of VTE in patients with pancreatic cancer (subdistribution hazard ratio per doubling of levels: 1.39 [95% confidence interval, 1.09-1.78], P = .007). Mice bearing PANC-1 tumors had increased levels of both active human and active mouse PAI-1 and decreased levels of plasmin activity. Importantly, mice bearing PANC-1 tumors exhibited impaired venous thrombus resolution 8 days after IVC stasis compared with nontumor controls. Our results suggest that PAI-1 contributes to VTE in pancreatic cancer.


Assuntos
Neoplasias Pancreáticas , Trombose Venosa , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Nus , Neoplasias Pancreáticas/complicações , Inibidor 1 de Ativador de Plasminogênio/genética , Trombose Venosa/etiologia
9.
J Thromb Haemost ; 18(11): 2899-2909, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33094904

RESUMO

BACKGROUND: The intrinsic pathway factors (F) XII and FXI have been shown to contribute to thrombosis in animal models. We assessed the role of FXII and FXI in venous thrombosis in three distinct mouse models. METHODS: Venous thrombosis was assessed in mice genetically deficient for either FXII or FXI. Three models were used: the inferior vena cava (IVC) stasis, IVC stenosis, and femoral vein electrolytic injury models. RESULTS: In the IVC stasis model, FXII and FXI deficiency did not affect the size of thrombi but their absence was associated with decreased levels of fibrin(ogen) and an increased level of the neutrophil extracellular trap marker citrullinated histone H3. In contrast, a deficiency of either FXII or FXI resulted in a significant and equivalent reduction in thrombus weight and incidence of thrombus formation in the IVC stenosis model. Thrombi formed in the IVC stenosis model contained significantly higher levels of citrullinated histone H3 compared with the thrombi formed in the IVC stasis model. Deletion of either FXII or FXI also resulted in a significant and equivalent reduction in both fibrin and platelet accumulation in the femoral vein electrolytic injury model. CONCLUSIONS: Collectively, these data indicate that FXII and FXI contribute to the size of venous thrombosis in models with blood flow and thrombus composition in a stasis model. This study also demonstrates the importance of using multiple mouse models to assess the role of a given protein in venous thrombosis.


Assuntos
Trombose , Trombose Venosa , Animais , Modelos Animais de Doenças , Fator XI/genética , Fator XII/genética , Fibrina , Camundongos , Trombose Venosa/genética
10.
Sci Rep ; 10(1): 17187, 2020 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-33057098

RESUMO

Heparan sulfate (HS) is a sulfated glycosaminoglycan abundant on the cell surface and in the extracellular matrix and has several biological activities including anticoagulation and anti-inflammation. Liver ischemia reperfusion injury is associated with coagulation and inflammatory responses. Here, we synthesized HS oligosaccharides with defined sulfation patterns and show that synthetic anticoagulant HS oligosaccharides limit liver ischemia reperfusion injury in a mouse model. Using a small targeted HS library, we demonstrate that an oligosaccharide that possesses both anticoagulant activity and binding affinity to HMGB1, the inflammatory target, decreases injury greater than oligosaccharides that only bind to HMGB1 or only have anticoagulant activity. HS oligosaccharides may represent a potential new therapeutic option for decreasing liver damage resulting from ischemia reperfusion injury.


Assuntos
Anticoagulantes/farmacologia , Heparitina Sulfato/farmacologia , Hepatopatias/tratamento farmacológico , Fígado/efeitos dos fármacos , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Coagulação Sanguínea/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Proteína HMGB1/metabolismo , Fígado/metabolismo , Hepatopatias/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oligossacarídeos/farmacologia , Traumatismo por Reperfusão/metabolismo
11.
Thromb Res ; 196: 200-205, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32896690

RESUMO

BACKGROUND: Plasma coagulation Factor XII (FXII) plays a crucial role in contact activation, ultimately regulating both the kinin-kallikrein system and the intrinsic pathway of coagulation. A growing body of evidence suggests that inhibition of FXII can prevent thrombosis. Given FXII does not appear to modulate hemostasis, targeting FXII is a promising strategy for the prevention of pathological thrombus formation without the hemostatic risks typically associated with anticoagulants. To this end, a subcutaneously administered investigational RNAi therapeutic targeting liver F12 mRNA (ALN-F12) was developed. AIM: To investigate the thrombo-protective and hemostatic effects of FXII reduction by ALN-F12 in rodent thrombosis and hemostasis models. METHODS: A single dose of ALN-F12 was subcutaneously administered to C57Bl/6 mice. After reaching steady state FXII reduction, the impact on thrombosis (ferric chloride arterial thrombosis and electrolytic injury induced venous thrombosis models) and hemostasis (saphenous vein injury and tail tip transection bleeding models) was evaluated. RESULT: Administration of ALN-F12 resulted in dose-dependent reductions of both liver F12 mRNA and plasma FXII protein. In mice, ALN-F12 led to dose-dependent reductions in platelet and fibrin accumulation in the venous electrolytic-injury model and in the time to occlusion in the ferric chloride arterial thrombosis model. At 10 mg/kg ALN-F12, the top dose level evaluated, this resulted in >95% reduction of FXII and ~10 fold reduction in fibrin deposition. Finally, hemostasis models showed that >95% reduction of FXII had no impact on bleeding time or blood loss. CONCLUSION: Our findings support that reduction of plasma Factor XII by ALN-F12 provided thrombo-protective effects with no increased bleeding risk in rodent models of thrombosis and hemostasis.


Assuntos
Deficiência do Fator XII , Hemostáticos , Trombose , Animais , Fator XII/genética , Hemostasia , Fígado , Camundongos , Camundongos Endogâmicos C57BL , RNA Interferente Pequeno , Trombose/genética , Trombose/prevenção & controle
12.
J Surg Res ; 254: 165-169, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32445932

RESUMO

BACKGROUND: Vein graft stenosis is a major complication of coronary artery bypass surgery and peripheral arterial bypass procedures. Experimental models of this clinical complication have used in vivo grafting procedures, relying on the relatively modest neointimal thickening in these models as a surrogate for clinical graft stenosis without regard to the donor site origin of the vein graft. MATERIALS AND METHODS: In a standard rat model of vein grafting, three different donor sites were used to supply veins used as interpositional grafts to the femoral artery: the superficial inferior epigastric vein, the common femoral vein, and the posterior facial vein (distal branch of the jugular vein). Grafts were harvested as 4 wk and histomorphometrically evaluated for the extent of neointimal formation and lumen narrowing. RESULTS: The posterior facial vein showed significantly thicker neointima and a greater extent of lumen narrowing than the other two graft sources, despite having a similar diameter to the femoral vein and nearly twice the initial diameter of the epigastric vein. CONCLUSIONS: The source of donor graft material can greatly influence the extent of neointimal response after interpositional vein grafting to arterial flow. These findings support use of the posterior facial vein graft over other more standard donor vein grafts in research directed at understanding the causes and prevention of vein graft stenosis.


Assuntos
Neointima/etiologia , Enxerto Vascular , Veias/transplante , Animais , Feminino , Ratos Endogâmicos Lew
13.
J Cardiovasc Dev Dis ; 7(1)2020 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-32197497

RESUMO

BACKGROUND: The timing for initiation of effective antithrombotic therapy relative to the onset of arterial thrombosis may influence outcomes. This report investigates the hypothesis that early administration of heparin anticoagulation relative to the onset of thrombotic occlusion will effect a reduction in occlusion. METHODS: A standard rat model of experimental thrombosis induction was used, injuring the carotid artery exposure with FeCl3-saturated filter paper, followed by flow monitoring for onset of occlusion and subsequent embolization events. Intravenous heparin administration (200 units/mL) was timed relative to the initiation of injury or onset of near occlusion, compared with controls (no heparin administration). RESULTS: No occlusion was found for delivery of heparin 5 min prior to thrombus induction, whereas all vessels occluded without heparin. Unstable (embolic) thrombi were seen with heparin given at or shortly after initial occlusion. Only 9% (1/11) of the vessels had permanent occlusion when heparin was given at the time of thrombotic onset (p < 0.0001 vs. unheparinized), while 50% occluded when heparin was delayed by 5 min (p > 0.05). CONCLUSIONS: These findings provide evidence that antithrombotic therapy may need to be administered prior to the onset of anticipated loss of patency, with less effectiveness when given after occlusion has occurred.

14.
Res Pract Thromb Haemost ; 4(1): 111-116, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31989092

RESUMO

BACKGROUND: The compositions of venous (red blood cell-rich) and arterial (platelet-rich) thrombi are mediated by distinct pathophysiologic processes; however, fibrin is a major structural component of both. The transglutaminase factor XIII (FXIII) stabilizes fibrin against mechanical and biochemical disruption and promotes red blood cell retention in contracted venous thrombi. Previous studies have shown factor XIII (FXIII) inhibition decreases whole blood clot mass and therefore, may be a therapeutic target for reducing venous thrombosis. The role of FXIII in arterial thrombogenesis is less studied, and the particular contribution of platelet FXIII remains unresolved. OBJECTIVE: To determine whether FXIII reduction prevents experimental arterial thrombogenesis. METHODS: Using wild-type mice and mice with genetically imposed deficiency in FXIII, we measured thrombus formation and stability following ferric chloride-induced arterial thrombosis. We also determined the impact of FXIII on the mass of contracted platelet-rich plasma clots. RESULTS: Following vessel injury, F13a+/+ , F13a+/- , and F13a-/- mice developed occlusive arterial thrombi. FXIII deficiency did not significantly reduce the incidence or prolong the time to occlusion. FXIII deficiency also did not alter the timing of reflow events or decrease platelet-rich clot mass. CONCLUSIONS: FXIII does not significantly alter the underlying pathophysiology of experimental arterial thrombus formation.

15.
Haematologica ; 105(1): 218-225, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31048354

RESUMO

Pancreatic cancer is associated with a high incidence of venous thromboembolism. Neutrophils have been shown to contribute to thrombosis in part by releasing neutrophil extracellular traps (NET). A recent study showed that increased plasma levels of the NET biomarker, citrullinated histone H3 (H3Cit), are associated with venous thromboembolism in patients with pancreatic and lung cancer but not in those with other types of cancer, including breast cancer. In this study, we examined the contribution of neutrophils and NET to venous thrombosis in nude mice bearing human pancreatic tumors. We found that tumor-bearing mice had increased circulating neutrophil counts and levels of granulocyte-colony stimulating factor, neutrophil elastase, H3Cit and cell-free DNA compared with controls. In addition, thrombi from tumor-bearing mice contained increased levels of the neutrophil marker Ly6G, as well as higher levels of H3Cit and cell-free DNA. Thrombi from tumor-bearing mice also had denser fibrin with thinner fibers consistent with increased thrombin generation. Importantly, either neutrophil depletion or administration of DNase I reduced the thrombus size in tumor-bearing but not in control mice. Our results, together with clinical data, suggest that neutrophils and NET contribute to venous thrombosis in patients with pancreatic cancer.


Assuntos
Armadilhas Extracelulares , Neoplasias Pancreáticas , Trombose Venosa , Animais , Humanos , Camundongos , Camundongos Nus , Neutrófilos , Trombose Venosa/etiologia
16.
Nat Commun ; 10(1): 4192, 2019 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-31519896

RESUMO

Lymph node (LN) metastases correspond with a worse prognosis in nearly all cancers, yet the occurrence of cancer spreading from LNs remains controversial. Additionally, the mechanisms explaining how cancers survive and exit LNs are largely unknown. Here, we show that breast cancer patients frequently have LN metastases that closely resemble distant metastases. In addition, using a microsurgical model, we show how LN metastasis development and dissemination is regulated by the expression of a chromatin modifier, histone deacetylase 11 (HDAC11). Genetic and pharmacologic blockade of HDAC11 decreases LN tumor growth, yet substantially increases migration and distant metastasis formation. Collectively, we reveal a mechanism explaining how HDAC11 plasticity promotes breast cancer growth as well as dissemination from LNs and suggest caution with the use of HDAC inhibitors.


Assuntos
Neoplasias da Mama/metabolismo , Histona Desacetilases/metabolismo , Linfonodos/metabolismo , Animais , Western Blotting , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Movimento Celular/fisiologia , Imunoprecipitação da Cromatina , Metilação de DNA/genética , Metilação de DNA/fisiologia , Citometria de Fluxo , Células HEK293 , Histona Desacetilases/genética , Humanos , Linfonodos/patologia , Metástase Linfática/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase em Tempo Real
17.
Blood Adv ; 2(21): 2848-2861, 2018 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-30381401

RESUMO

Dyslipidemia is a risk factor for clinically significant thrombotic events. In this condition, scavenger receptor CD36 potentiates platelet reactivity through recognition of circulating oxidized lipids. CD36 promotes thrombosis by activating redox-sensitive signaling molecules, such as the MAPK extracellular signal-regulated kinase 5 (ERK5). However, the events downstream of platelet ERK5 are not clear. In this study, we report that oxidized low-density lipoprotein (oxLDL) promotes exposure of procoagulant phosphatidylserine (PSer) on platelet surfaces. Studies using pharmacologic inhibitors indicate that oxLDL-CD36 interaction-induced PSer exposure requires apoptotic caspases in addition to the downstream CD36-signaling molecules Src kinases, hydrogen peroxide, and ERK5. Caspases promote PSer exposure and, subsequently, recruitment of the prothrombinase complex, resulting in the generation of fibrin from the activation of thrombin. Caspase activity was observed when platelets were stimulated with oxLDL. This was prevented by inhibiting CD36 and ERK5. Furthermore, oxLDL potentiates convulxin/glycoprotein VI-mediated fibrin formation by platelets, which was prevented when CD36, ERK5, and caspases were inhibited. Using 2 in vivo arterial thrombosis models in apoE-null hyperlipidemic mice demonstrated enhanced arterial fibrin accumulation upon vessel injury. Importantly, absence of ERK5 in platelets or mice lacking CD36 displayed decreased fibrin accumulation in high-fat diet-fed conditions comparable to that seen in chow diet-fed animals. These findings suggest that platelet signaling through CD36 and ERK5 induces a procoagulant phenotype in the hyperlipidemic environment by enhancing caspase-mediated PSer exposure.


Assuntos
Plaquetas/metabolismo , Antígenos CD36/metabolismo , Caspases/metabolismo , Fibrina/metabolismo , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Fosfatidilserinas/metabolismo , Animais , Plaquetas/citologia , Antígenos CD36/antagonistas & inibidores , Venenos de Crotalídeos/farmacologia , Dieta Hiperlipídica , Modelos Animais de Doenças , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Humanos , Hiperlipidemias/complicações , Hiperlipidemias/patologia , Lectinas Tipo C , Lipoproteínas LDL/farmacologia , Camundongos , Camundongos Knockout , Proteína Quinase 7 Ativada por Mitógeno/antagonistas & inibidores , Ativação Plaquetária/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Trombose/etiologia , Trombose/patologia , Quinases da Família src/metabolismo
18.
Blood Coagul Fibrinolysis ; 29(7): 636-643, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30113321

RESUMO

: The murine FeCl3 model is a widely used model for studying arterial thrombosis, yet provides limited information from each mouse, often only a single time point for the onset of occlusion (defined as the time to occlusion; TTO). To optimize data from the murine ferric chloride model of thrombosis. FeCl3 injury was induced in the carotid arteries of wild-type and Factor IX (FIX) knockout mice, with infusion of recombinant FIX (rFIX) to normalize FIX deficiency at various times around FeCl3 injury. The TTO was recorded as a percentage of baseline flow as occlusion continued to zero flow, with identification of reflow events. The TTO among the treatment groups of FIX-deficient mice showed no statistical differences, except with physiological saline-treated FIX-deficient mice and those receiving delayed treatment. Incidences of occlusion were 100% for wild-type mice and FIX-deficient mice receiving slow infusions of rFIX at early times around the FeCl3 application. In contrast, only 68% of FIX-deficient mice achieved occlusion with preinfusion of rFIX and none occluded with delayed rFIX infusion. A majority of occluded vessels exhibited reflow events, with significantly lower incidence for slow infusion of rFIX starting 4 min after FeCl3 application in comparison with preinjury bolus, demonstrating characterization of a differential response to timing and infusion rates of treatment. Simple use of the time to occlusion may not maximize data available from the FeCl3 arterial thrombosis model. Inclusion of documenting reflow events can extend the useful data obtained with application of this model.


Assuntos
Fator IX/administração & dosagem , Trombose/tratamento farmacológico , Animais , Artérias Carótidas/patologia , Cloretos , Compostos Férricos , Hemofilia B/tratamento farmacológico , Camundongos , Camundongos Knockout , Trombose/induzido quimicamente , Resultado do Tratamento
19.
Arterioscler Thromb Vasc Biol ; 38(4): 816-828, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29419409

RESUMO

OBJECTIVE: PS (protein S) is a plasma protein that directly inhibits the coagulation FIXa (factor IXa) in vitro. Because elevated FIXa is associated with increased risk of venous thromboembolism, it is important to establish how PS inhibits FIXa function in vivo. The goal of this study is to confirm direct binding of PS with FIXa in vivo, identify FIXa amino acid residues required for binding PS in vivo, and use an enzymatically active FIXa mutant that is unable to bind PS to measure the significance of PS-FIXa interaction in hemostasis. APPROACH AND RESULTS: We demonstrate that PS inhibits FIXa in vivo by associating with the FIXa heparin-binding exosite. We used fluorescence tagging, immunohistochemistry, and protein-protein crosslinking to show in vivo interaction between FIXa and PS. Importantly, platelet colocalization required a direct interaction between the 2 proteins. FIXa and PS also coimmunoprecipitated from plasma, substantiating their interaction in a physiological milieu. PS binding to FIXa and PS inhibition of the intrinsic Xase complex required residues K132, K126, and R170 in the FIXa heparin-binding exosite. A double mutant, K132A/R170A, retained full activity but could not bind to PS. Crucially, Hemophilia B mice infused with FIXa K132A/R170A displayed an accelerated rate of fibrin clot formation compared with wild-type FIXa. CONCLUSIONS: Our findings establish PS as an important in vivo inhibitor of FIXa. Disruption of the interaction between PS and FIXa causes an increased rate of thrombus formation in mice. This newly discovered function of PS implies an unexploited target for antithrombotic therapeutics.


Assuntos
Plaquetas/metabolismo , Fator IXa/metabolismo , Hemofilia B/sangue , Hemostasia , Heparina/metabolismo , Proteína S/metabolismo , Trombose Venosa/prevenção & controle , Animais , Sítios de Ligação , Ligação Competitiva , Coagulantes/administração & dosagem , Modelos Animais de Doenças , Fator IX/genética , Fator IX/metabolismo , Fator IXa/administração & dosagem , Fator IXa/genética , Hemofilia B/tratamento farmacológico , Hemofilia B/genética , Hemostasia/efeitos dos fármacos , Humanos , Infusões Intravenosas , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Trombose Venosa/sangue , Trombose Venosa/genética
20.
Blood Adv ; 2(1): 25-35, 2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29344582

RESUMO

The transglutaminase factor XIII (FXIII) stabilizes clots against mechanical and biochemical disruption and is essential for hemostasis. In vitro and in vivo models of venous thrombosis demonstrate that FXIII mediates clot size by promoting red blood cell (RBC) retention. However, the key source of FXIII and whether FXIII activity can be reduced to suppress thrombosis without imposing deleterious hemostatic consequences are 2 critical unresolved questions. FXIII is present in multiple compartments, including plasma (FXIIIplasma) as a heterotetramer of A2 and B2 subunits and platelets (FXIIIplt) as an A2 homodimer. We determined the role of the FXIII compartment and level in clot contraction, composition, and size in vitro and using in vivo models of hemostasis and venous thrombosis. Reducing overall FXIII levels decreased whole blood clot weight but did not alter thrombin generation or contraction of platelet-rich plasma clots. In reconstituted platelet-rich plasma and whole blood clot contraction assays, FXIIIplasma, but not FXIIIplt, produced high-molecular-weight fibrin crosslinks, promoted RBC retention, and increased clot weights. Genetically imposed reduction of FXIII delayed FXIII activation and fibrin crosslinking, suggesting FXIII levels mediate the kinetics of FXIII activation and activity and that the timing of these processes is a critical determinant of RBC retention during clot formation and contraction. A 50% reduction in FXIIIplasma produced significantly smaller venous thrombi but did not increase bleeding in tail transection or saphenous vein puncture models in vivo. Collectively, these findings suggest that partial FXIII reduction may be a therapeutic strategy for reducing venous thrombosis.


Assuntos
Eritrócitos/patologia , Fator XIII/fisiologia , Trombose/patologia , Trombose Venosa/patologia , Animais , Plaquetas , Fibrina/metabolismo , Hemorragia/etiologia , Camundongos , Plasma/química , Trombina/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...