Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Intervalo de año de publicación
1.
bioRxiv ; 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38352439

RESUMEN

While evolution is often considered from a DNA- and protein-centric view, RNA-based regulation can also impact gene expression and protein sequences. Here we examined interspecies differences in RNA-protein interactions using the conserved neuronal RNA binding protein, Unkempt (UNK) as model. We find that roughly half of mRNAs bound in human are also bound in mouse. Unexpectedly, even when transcript-level binding was conserved across species differential motif usage was prevalent. To understand the biochemical basis of UNK-RNA interactions, we reconstituted the human and mouse UNK-RNA interactomes using a high-throughput biochemical assay. We uncover detailed features driving binding, show that in vivo patterns are captured in vitro, find that highly conserved sites are the strongest bound, and associate binding strength with downstream regulation. Furthermore, subtle sequence differences surrounding motifs are key determinants of species-specific binding. We highlight the complex features driving protein-RNA interactions and how these evolve to confer species-specific regulation.

2.
Blood ; 143(5): 444-455, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-37883802

RESUMEN

ABSTRACT: Transglutaminase factor XIII (FXIII) is essential for hemostasis, wound healing, and pregnancy maintenance. Plasma FXIII is composed of A and B subunit dimers synthesized in cells of hematopoietic origin and hepatocytes, respectively. The subunits associate tightly in circulation as FXIII-A2B2. FXIII-B2 stabilizes the (pro)active site-containing FXIII-A subunits. Interestingly, people with genetic FXIII-A deficiency have decreased FXIII-B2, and therapeutic infusion of recombinant FXIII-A2 (rFXIII-A2) increases FXIII-B2, suggesting FXIII-A regulates FXIII-B secretion, production, and/or clearance. We analyzed humans and mice with genetic FXIII-A deficiency and developed a mouse model of rFXIII-A2 infusion to define mechanisms mediating plasma FXIII-B levels. Like humans with FXIII-A deficiency, mice with genetic FXIII-A deficiency had reduced circulating FXIII-B2, and infusion of FXIII-A2 increased FXIII-B2. FXIII-A-deficient mice had normal hepatic function and did not store FXIII-B in liver, indicating FXIII-A does not mediate FXIII-B secretion. Transcriptional analysis and polysome profiling indicated similar F13b levels and ribosome occupancy in FXIII-A-sufficient and -deficient mice and in FXIII-A-deficient mice infused with rFXIII-A2, indicating FXIII-A does not induce de novo FXIII-B synthesis. Unexpectedly, pharmacokinetic/pharmacodynamic modeling of FXIII-B antigen after rFXIII-A2 infusion in humans and mice suggested FXIII-A2 slows FXIII-B2 loss from plasma. Accordingly, comparison of free FXIII-B2 vs FXIII-A2-complexed FXIII-B2 (FXIII-A2B2) infused into mice revealed faster clearance of free FXIII-B2. These data show FXIII-A2 prevents FXIII-B2 loss from circulation and establish the mechanism underlying FXIII-B2 behavior in FXIII-A deficiency and during rFXIII-A2 therapy. Our findings reveal a unique, reciprocal relationship between independently synthesized subunits that mediate an essential hemostatic protein in circulation. This trial was registered at www.ClinicalTrials.com as #NCT00978380.


Asunto(s)
Deficiencia del Factor XIII , Animales , Femenino , Humanos , Ratones , Embarazo , Pruebas de Coagulación Sanguínea , Factor XIII/metabolismo , Deficiencia del Factor XIII/genética , Factor XIIIa/genética , Hemostasis , Hemostáticos/sangre
3.
J Thromb Haemost ; 20(7): 1674-1683, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35466511

RESUMEN

BACKGROUND: Factor VIII (FVIII) binding to endogenous von Willebrand factor (VWF) has constrained half-life extension of recombinant FVIII (rFVIII) products for hemophilia A. Efanesoctocog alfa (rFVIIIFc-VWF-XTEN; BIVV001) is a novel fusion protein designed to decouple FVIII from VWF in circulation and maximize half-life prolongation by XTEN® polypeptides and Fc fusion. FVIII, VWF, and platelets interact to achieve normal hemostasis. Thus, bioengineered FVIII replacement products, such as efanesoctocog alfa, require comprehensive assessment of their hemostatic potential. OBJECTIVES: We compared functional clot formation and injury-induced platelet accumulation between efanesoctocog alfa and rFVIII. PATIENTS/METHODS: The hemostatic potential of efanesoctocog alfa and rFVIII were assessed by measuring their dose-dependent effects on in vitro fibrin generation in hemophilic plasma and in vivo injury-induced platelet accumulation using intravital microscopy and repeat saphenous vein laser-induced injuries in hemophilia A mice. RESULTS: Equal concentrations of efanesoctocog alfa or rFVIII (up to 1 IU/ml) added to plasma from patients with hemophilia A elicited similar kinetics for dose-dependent fibrin polymerization between factor products. In the presence of tissue plasminogen activator (tPA), clots formed had similar stability between products. Single intravenous doses (50, 100, or 150 IU/kg) of efanesoctocog alfa or rFVIII shortly before repeat saphenous vein laser-induced injuries increased platelet accumulation over time in a dose-dependent manner in hemophilia A mice. Platelet deposition kinetics were similar between products. CONCLUSIONS: Equivalent doses of efanesoctocog alfa and rFVIII had similar efficacy in promoting fibrin clot formation and injury-induced platelet accumulation. The hemostatic potential of efanesoctocog alfa was indistinguishable from that of rFVIII.


Asunto(s)
Hemofilia A , Hemostáticos , Animales , Factor VIII/metabolismo , Fibrina , Hemostáticos/uso terapéutico , Humanos , Ratones , Activador de Tejido Plasminógeno/uso terapéutico , Factor de von Willebrand/metabolismo
4.
Life Sci Alliance ; 5(5)2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35173014

RESUMEN

Cyclin E/CDK2 drives cell cycle progression from G1 to S phase. Despite the toxicity of cyclin E overproduction in mammalian cells, the cyclin E gene is overexpressed in some cancers. To further understand how cells can tolerate high cyclin E, we characterized non-transformed epithelial cells subjected to chronic cyclin E overproduction. Cells overproducing cyclin E, but not cyclins D or A, briefly experienced truncated G1 phases followed by a transient period of DNA replication origin underlicensing, replication stress, and impaired proliferation. Individual cells displayed substantial intercellular heterogeneity in cell cycle dynamics and CDK activity. Each phenotype improved rapidly despite high cyclin E-associated activity. Transcriptome analysis revealed adapted cells down-regulated a cohort of G1-regulated genes. Withdrawing cyclin E from adapted cells only partially reversed underlicensing indicating that adaptation is at least partly non-genetic. This study provides evidence that mammalian cyclin E/CDK inhibits origin licensing indirectly through premature S phase onset and provides mechanistic insight into the relationship between CDKs and licensing. It serves as an example of oncogene adaptation that may recapitulate molecular changes during tumorigenesis.


Asunto(s)
Ciclina E/genética , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina/genética , Animales , Ciclo Celular , División Celular , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Replicación del ADN , Fase G1 , Expresión Génica/genética , Regulación de la Expresión Génica/genética , Humanos , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Fase S
5.
J Clin Invest ; 131(22)2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34779407

RESUMEN

High expression of LIN28B is associated with aggressive malignancy and poor survival. Here, probing MYCN-amplified neuroblastoma as a model system, we showed that LIN28B expression was associated with enhanced cell migration in vitro and invasive and metastatic behavior in murine xenografts. Sequence analysis of the polyribosome fraction of LIN28B-expressing neuroblastoma cells revealed let-7-independent enrichment of transcripts encoding components of the translational and ribosomal apparatus and depletion of transcripts of neuronal developmental programs. We further observed that LIN28B utilizes both its cold shock and zinc finger RNA binding domains to preferentially interact with MYCN-induced transcripts of the ribosomal complex, enhancing their translation. These data demonstrated that LIN28B couples the MYCN-driven transcriptional program to enhanced ribosomal translation, thereby implicating LIN28B as a posttranscriptional driver of the metastatic phenotype.


Asunto(s)
Proteína Proto-Oncogénica N-Myc/fisiología , Metástasis de la Neoplasia , Neuroblastoma/patología , Proteínas de Unión al ARN/fisiología , Ribosomas/fisiología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Neuroblastoma/etiología
6.
Nat Commun ; 12(1): 1569, 2021 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-33692367

RESUMEN

Quantitative criteria to identify proteins as RNA-binding proteins (RBPs) are presently lacking, as are criteria to define RBP target RNAs. Here, we develop an ultraviolet (UV) cross-linking immunoprecipitation (CLIP)-sequencing method, easyCLIP. easyCLIP provides absolute cross-link rates, as well as increased simplicity, efficiency, and capacity to visualize RNA libraries during sequencing library preparation. Measurement of >200 independent cross-link experiments across >35 proteins identifies an RNA cross-link rate threshold that distinguishes RBPs from non-RBPs and defines target RNAs as those with a complex frequency unlikely for a random protein. We apply easyCLIP to the 33 most recurrent cancer mutations across 28 RBPs, finding increased RNA binding per RBP molecule for KHDRBS2 R168C, A1CF E34K and PCBP1 L100P/Q cancer mutations. Quantitating RBP-RNA interactions can thus nominate proteins as RBPs and define the impact of specific disease-associated RBP mutations on RNA association.


Asunto(s)
Proteínas de Unión al ARN/química , ARN/química , Animales , Sitios de Unión , Humanos , Inmunoprecipitación , ARN/metabolismo , ARN/efectos de la radiación , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/efectos de la radiación , Rayos Ultravioleta
7.
J Thromb Haemost ; 17(7): 1044-1052, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30887655

RESUMEN

Essentials Non-factor VIII (FVIII) therapies for hemophilia A, such as bispecific antibodies (bsAbs), are in development. Bispecific antibodies are intrinsically different from FVIII and lack many of the same regulatory mechanisms. These differences complicate assignment and interpretation of FVIII-equivalent activity. Inability to assign FVIII equivalence compromises our capacity to assess hemostatic potential of bsAb therapies. BACKGROUND: Activated factor VIII (FVIIIa) mimetic bsAbs aim to enable prophylactic treatment of hemophilia A patients with and without inhibitors. With different mechanisms of action, benchmarking their activity against FVIII to determine efficacious yet safe dosage is difficult. OBJECTIVE: To compare the activities of sequence identical emicizumab (SI-Emi) and another bsAb, BS-027125, to recombinant FVIII (rFVIII) using clinical and nonclinical assays and to evaluate our ability to assign a FVIII-equivalent value to bsAbs and implications thereof. METHODS: Activities of SI-Emi, BS-027125, and rFVIII were measured by one-stage clotting assay, chromogenic factor Xa generation assay, and thrombin generation assay. We also assessed the activity of anti-FIXa and anti-FX bivalent homodimers of each bsAb and probed the effect of different reagents in thrombin generation assay (TGA). RESULTS: The FVIII-like activity of SI-Emi and BS-027125 ranged greatly across each assay, varying both by parameter measured within an assay and by reagents used. Notably, SI-Emi anti-FIXa bivalent homodimer had meaningful activity in several assays, whereas BS-027125 anti-FIXa bivalent homodimer only had activity in the chromogenic assay. Surprisingly, SI-Emi displayed activity in the absence of phospholipids, while BS-027125 had minimal phospholipid-independent activity. CONCLUSIONS: Bispecific antibodies demonstrate little consistency between assays tested here owing to intrinsic differences between FVIII and bsAbs. While some trends are shared, the bsAbs also differ in mechanism. These inconsistencies complicate assignment of FVIII-equivalent values to bsAbs. Ultimately, a deeper mechanistic understanding of bsAbs as well as bsAb-tailored assays are needed to monitor and predict their hemostatic potential and long-term efficacy and safety confidently.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Anticuerpos Monoclonales Humanizados/farmacología , Mimetismo Biológico , Factor VIII/farmacología , Hematínicos/farmacología , Hemofilia A/tratamiento farmacológico , Hemostasis/efectos de los fármacos , Equivalencia Terapéutica , Pruebas de Coagulación Sanguínea , Factor VIII/inmunología , Factor Xa/metabolismo , Hemofilia A/sangre , Hemofilia A/diagnóstico , Hemofilia A/inmunología , Humanos , Resonancia por Plasmón de Superficie , Trombina/metabolismo
9.
Respir Res ; 17(1): 89, 2016 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-27450419

RESUMEN

UNLABELLED: Sulforaphane (SFN), a naturally occurring isothiocyanate found in cruciferous vegetables, is implicated as a possible therapy for airway inflammation via induction of the transcription factor NF-E2-related factor 2 (NRF2). In this proof-of-concept clinical study, we show that supplementation of SFN with broccoli sprout homogenate in healthy human subjects did not induce expression of antioxidant genes or protect against neutrophilic airway inflammation in an ozone-exposure model. Therefore, dietary sulforaphane supplementation is not a promising candidate for larger scale clinical trials targeting airway inflammation. TRIAL REGISTRATION: NCT01625130 . Registered 19 June, 2012.


Asunto(s)
Antiinflamatorios/uso terapéutico , Isotiocianatos/uso terapéutico , Trastornos Leucocíticos/prevención & control , Pulmón/efectos de los fármacos , Factor 2 Relacionado con NF-E2/agonistas , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Neumonía/prevención & control , Adolescente , Adulto , Antiinflamatorios/aislamiento & purificación , Brassica/química , Estudios Cruzados , Femenino , Voluntarios Sanos , Humanos , Isotiocianatos/aislamiento & purificación , Trastornos Leucocíticos/inducido químicamente , Trastornos Leucocíticos/inmunología , Trastornos Leucocíticos/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Masculino , Persona de Mediana Edad , Factor 2 Relacionado con NF-E2/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Ozono , Fitoterapia , Plantas Medicinales , Neumonía/inducido químicamente , Neumonía/inmunología , Neumonía/metabolismo , Sulfóxidos , Adulto Joven
10.
EBioMedicine ; 5: 175-82, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27077125

RESUMEN

Thrombus formation leading to vaso-occlusive events is a major cause of death, and involves complex interactions between coagulation, fibrinolytic and innate immune systems. Leukocyte recruitment is a key step, mediated partly by chemotactic complement activation factors C3a and C5a. However, mechanisms mediating C3a/C5a generation during thrombosis have not been studied. In a murine venous thrombosis model, levels of thrombin-antithrombin complexes poorly correlated with C3a and C5a, excluding a central role for thrombin in C3a/C5a production. However, clot weight strongly correlated with C5a, suggesting processes triggered during thrombosis promote C5a generation. Since thrombosis elicits fibrinolysis, we hypothesized that plasmin activates C5 during thrombosis. In vitro, the catalytic efficiency of plasmin-mediated C5a generation greatly exceeded that of thrombin or factor Xa, but was similar to the recognized complement C5 convertases. Plasmin-activated C5 yielded a functional membrane attack complex (MAC). In an arterial thrombosis model, plasminogen activator administration increased C5a levels. Overall, these findings suggest plasmin bridges thrombosis and the immune response by liberating C5a and inducing MAC assembly. These new insights may lead to the development of strategies to limit thrombus formation and/or enhance resolution.


Asunto(s)
Arterias/inmunología , Complemento C5a/inmunología , Fibrinolisina/inmunología , Trombosis de la Vena/inmunología , Animales , Antitrombina III/efectos de los fármacos , Antitrombina III/inmunología , Arterias/efectos de los fármacos , Arterias/patología , Activación de Complemento/efectos de los fármacos , Activación de Complemento/inmunología , Complemento C3a/biosíntesis , Complemento C3a/inmunología , Complemento C5a/biosíntesis , Complejo de Ataque a Membrana del Sistema Complemento/efectos de los fármacos , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Factor Xa/inmunología , Factor Xa/metabolismo , Fibrinolisina/metabolismo , Humanos , Ratones , Péptido Hidrolasas/efectos de los fármacos , Péptido Hidrolasas/inmunología , Activadores Plasminogénicos/administración & dosificación , Trombina/inmunología , Trombina/metabolismo , Trombosis de la Vena/tratamiento farmacológico , Trombosis de la Vena/patología
12.
PLoS One ; 10(4): e0124448, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25915545

RESUMEN

Transglutaminases are a superfamily of isoenzymes found in cells and plasma. These enzymes catalyze the formation of ε-N-(γ-glutamyl)-lysyl crosslinks between proteins. Cystamine blocks transglutaminase activity and is used in vitro in human samples and in vivo in mice and rats in studies of coagulation, immune dysfunction, and inflammatory disease. These studies have suggested cystamine blocks fibrin crosslinking and has anti-inflammatory effects, implicating transglutaminase activity in the pathogenesis of several diseases. We measured the effects of cystamine on fibrin crosslinking, tissue factor-triggered plasma clot formation and thrombin generation, and coagulation factor enzymatic activity. At concentrations that blocked fibrin crosslinking, cystamine also inhibited plasma clot formation and reduced thrombin generation. Cystamine inhibited the amidolytic activity of coagulation factor XI and thrombin towards chromogenic substrates. These findings demonstrate that cystamine exhibits anticoagulant activity during coagulation. Given the close relationship between coagulation and inflammation, these findings suggest prior studies that used cystamine to implicate transglutaminase activity in disease pathogenesis warrant re-examination.


Asunto(s)
Anticoagulantes/química , Anticoagulantes/farmacología , Cistamina/química , Cistamina/farmacología , Animales , Coagulación Sanguínea/efectos de los fármacos , Factor XIa/metabolismo , Fibrina/metabolismo , Humanos , Ratones , Ratas , Trombina , Transglutaminasas/antagonistas & inhibidores , Transglutaminasas/metabolismo
14.
J Allergy Clin Immunol ; 135(2): 379-85, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25195169

RESUMEN

BACKGROUND: Asthma with neutrophil predominance is challenging to treat with corticosteroids. Novel treatment options for asthma include those that target innate immune activity. Recent literature has indicated a significant role for IL-1ß in both acute and chronic neutrophilic asthma. OBJECTIVE: This study used inhaled endotoxin (LPS) challenge as a model of innate immune activation to (1) assess the safety of the IL-1 receptor antagonist anakinra in conjunction with inhaled LPS and (2) to test the hypothesis that IL-1 blockade will suppress the acute neutrophil response to challenge with inhaled LPS. METHODS: In a phase I clinical study 17 healthy volunteers completed a double-blind, placebo-controlled crossover study in which they received 2 daily subcutaneous doses of 1 mg/kg anakinra (maximum dose, 100 mg) or saline (placebo). One hour after the second treatment dose, subjects underwent an inhaled LPS challenge. Induced sputum was assessed for neutrophils 4 hours after inhaled LPS. The effect of anakinra compared with placebo on airway neutrophil counts and airway proinflammatory cytokine levels after LPS challenge was compared by using a linear mixed-model approach. RESULTS: Anakinra pretreatment significantly diminished airway neutrophilia compared with placebo. LPS-induced IL-1ß, IL-6, and IL-8 levels were significantly reduced during the anakinra treatment period compared with those seen after placebo. Subjects tolerated the anakinra treatment well without an increased frequency of infections attributable to anakinra treatment. CONCLUSIONS: Anakinra effectively reduced airway neutrophilic inflammation and resulted in no serious adverse events in a model of inhaled LPS challenge. Anakinra is a potential therapeutic candidate for treatment of asthma with neutrophil predominance in diseased populations.


Asunto(s)
Antiinflamatorios/uso terapéutico , Endotoxinas/efectos adversos , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Receptores de Interleucina-1/antagonistas & inhibidores , Enfermedades Respiratorias/inducido químicamente , Enfermedades Respiratorias/tratamiento farmacológico , Administración por Inhalación , Adulto , Antiinflamatorios/administración & dosificación , Estudios Cruzados , Citocinas/metabolismo , Endotoxinas/administración & dosificación , Femenino , Voluntarios Sanos , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Proteína Antagonista del Receptor de Interleucina 1/administración & dosificación , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Recuento de Leucocitos , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/efectos adversos , Masculino , Infiltración Neutrófila/efectos de los fármacos , Infiltración Neutrófila/inmunología , Premedicación , Receptores de Interleucina-1/metabolismo , Enfermedades Respiratorias/inmunología , Enfermedades Respiratorias/metabolismo , Esputo/citología , Esputo/inmunología , Esputo/metabolismo , Resultado del Tratamiento , Adulto Joven
15.
J Clin Invest ; 124(8): 3590-600, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24983320

RESUMEN

Venous thrombi, fibrin- and rbc-rich clots triggered by inflammation and blood stasis, underlie devastating, and sometimes fatal, occlusive events. During intravascular fibrin deposition, rbc are thought to become passively trapped in thrombi and therefore have not been considered a modifiable thrombus component. In the present study, we determined that activity of the transglutaminase factor XIII (FXIII) is critical for rbc retention within clots and directly affects thrombus size. Compared with WT mice, mice carrying a homozygous mutation in the fibrinogen γ chain (Fibγ390-396A) had a striking 50% reduction in thrombus weight due to reduced rbc content. Fibrinogen from mice harboring the Fibγ390-396A mutation exhibited reduced binding to FXIII, and plasma from these mice exhibited delayed FXIII activation and fibrin crosslinking, indicating these residues mediate FXIII binding and activation. FXIII-deficient mice phenocopied mice carrying Fibγ390-396A and produced smaller thrombi with fewer rbc than WT mice. Importantly, FXIII-deficient human clots also exhibited reduced rbc retention. The addition of FXIII to FXIII-deficient clots increased rbc retention, while inhibition of FXIII activity in normal blood reduced rbc retention and produced smaller clots. These findings establish the FXIII-fibrinogen axis as a central determinant in venous thrombogenesis and identify FXIII as a potential therapeutic target for limiting venous thrombosis.


Asunto(s)
Eritrocitos/metabolismo , Eritrocitos/patología , Factor XIII/metabolismo , Fibrinógeno/genética , Fibrinógeno/metabolismo , Trombosis de la Vena/sangre , Animales , Coagulación Sanguínea , Modelos Animales de Enfermedad , Deficiencia del Factor XIII/sangre , Deficiencia del Factor XIII/genética , Factor XIIIa/genética , Factor XIIIa/metabolismo , Homocigoto , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Proteínas Mutantes/genética , Mutación , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Trombosis de la Vena/genética
16.
Thromb Res ; 133 Suppl 1: S38-40, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24759140

RESUMEN

Deep vein thrombosis and pulmonary embolism, collectively termed venous thromboembolism (VTE), affect over 1 million Americans each year. VTE is triggered by inflammation and blood stasis leading to the formation of thrombi rich in fibrin and red blood cells (RBCs). However, little is known about mechanisms regulating fibrin and RBC incorporation into venous thrombi, or how these components mediate thrombus size or resolution. Both elevated circulating fibrinogen (hyperfibrinogenemia) and abnormal fibrin(ogen) structure and function, including increased fibrin network density and resistance to fibrinolysis, have been observed in plasmas from patients with VTE. Abnormalities in RBC number and/or function have also been associated with VTE risk. RBC contributions to VTE are thought to stem from their effects on blood viscosity and margination of platelets to the vessel wall. More recent studies suggest RBCs also express phosphatidylserine, support thrombin generation, and decrease fibrinolysis. RBC interactions with fibrin(ogen) and cells, including platelets and endothelial cells, may also promote thrombus formation. The contributions of fibrin(ogen) and RBCs to the pathophysiology of VTE warrants further investigation.


Asunto(s)
Eritrocitos/patología , Fibrinógeno/metabolismo , Venas/patología , Trombosis de la Vena/patología , Animales , Coagulación Sanguínea , Eritrocitos/metabolismo , Fibrinógeno/análisis , Humanos , Venas/metabolismo , Trombosis de la Vena/sangre , Trombosis de la Vena/metabolismo
18.
Arterioscler Thromb Vasc Biol ; 33(8): 1829-36, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23723374

RESUMEN

OBJECTIVE: Individuals with elevated prothrombin, including those with the prothrombin G20210A mutation, have increased risk of venous thrombosis. Although these individuals do not have increased circulating prothrombotic biomarkers, their plasma demonstrates increased tissue factor-dependent thrombin generation in vitro. The objectives of this study were to determine the pathological role of elevated prothrombin in venous and arterial thrombosis in vivo, and distinguish thrombogenic mechanisms in these vessels. APPROACH AND RESULTS: Prothrombin was infused into mice to raise circulating levels. Venous thrombosis was induced by electrolytic stimulus to the femoral vein or inferior vena cava ligation. Arterial thrombosis was induced by electrolytic stimulus or ferric chloride application to the carotid artery. Mice infused with prothrombin demonstrated increased tissue factor-triggered thrombin generation measured ex vivo, but did not have increased circulating prothrombotic biomarkers in the absence of vessel injury. After venous injury, elevated prothrombin increased thrombin generation and the fibrin accumulation rate and total amount of fibrin ≈ 3-fold, producing extended thrombi with increased mass. However, elevated prothrombin did not accelerate platelet accumulation, increase the fibrin accumulation rate, or shorten the vessel occlusion time after arterial injury. CONCLUSIONS: These findings reconcile previously discordant findings on thrombin generation in hyperprothrombinemic individuals measured ex vivo and in vitro, and show elevated prothrombin promotes venous, but not arterial, thrombosis in vivo.


Asunto(s)
Coagulación Sanguínea/fisiología , Protrombina/metabolismo , Trombofilia/metabolismo , Trombosis de la Vena/metabolismo , Animales , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/efectos de los fármacos , Plaquetas/fisiología , Arterias Carótidas/fisiología , Cloruros/farmacología , Modelos Animales de Enfermedad , Vena Femoral/fisiología , Compuestos Férricos/farmacología , Fibrina/metabolismo , Humanos , Ratones , Noxas/farmacología , Protrombina/farmacología , Factores de Riesgo , Trombofilia/inducido químicamente , Trombofilia/epidemiología , Vena Cava Inferior/fisiología , Trombosis de la Vena/inducido químicamente , Trombosis de la Vena/epidemiología
19.
Blood ; 119(23): 5543-52, 2012 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-22547577

RESUMEN

Cancer patients often have an activated clotting system and are at increased risk for venous thrombosis. In the present study, we analyzed tissue factor (TF) expression in 4 different human pancreatic tumor cell lines for the purpose of producing derivative tumors in vivo. We found that 2 of the lines expressed TF and released TF-positive microparticles (MPs) into the culture medium. The majority of TF protein in the culture medium was associated with MPs. Only TF-positive cell lines activated coagulation in nude mice, and this activation was abolished by an anti-human TF Ab. Of the 2 TF-positive lines, only one produced detectable levels of human MP TF activity in the plasma when grown orthotopically in nude mice. Surprisingly, < 5% of human TF protein in plasma from tumor-bearing mice was associated with MPs. Mice with TF-positive tumors and elevated levels of circulating TF-positive MPs had increased thrombosis in a saphenous vein model. In contrast, we observed no difference in thrombus weight between tumor-bearing and control mice in an inferior vena cava stenosis model. The results of the present study using a xenograft mouse model suggest that tumor TF activates coagulation, whereas TF on circulating MPs may trigger venous thrombosis.


Asunto(s)
Coagulación Sanguínea , Neoplasias Pancreáticas/complicaciones , Neoplasias Pancreáticas/metabolismo , Tromboplastina/metabolismo , Trombosis de la Vena/complicaciones , Animales , Línea Celular Tumoral , Micropartículas Derivadas de Células/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Regulación Neoplásica de la Expresión Génica , Hemostasis , Humanos , Ratones , Ratones Desnudos , Neoplasias Pancreáticas/genética , ARN Mensajero/genética , Tromboplastina/genética , Trombosis de la Vena/metabolismo
20.
Anesth Analg ; 114(2): 275-85, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22104070

RESUMEN

Virchow's triad is traditionally invoked to explain pathophysiologic mechanisms leading to thrombosis, alleging concerted roles for abnormalities in blood composition, vessel wall components, and blood flow in the development of arterial and venous thrombosis. Given the tissue-specific bleeding observed in hemophilia patients, it may be instructive to consider the principles of Virchow's triad when investigating mechanisms operant in hemostatic disorders as well. Blood composition (the function of circulating blood cells and plasma proteins) is the most well studied component of the triad. For example, increased levels of plasma procoagulant proteins such as prothrombin and fibrinogen are established risk factors for thrombosis, whereas deficiencies in plasma factors VIII and IX result in bleeding (hemophilia A and B, respectively). Vessel wall (cellular) components contribute adhesion molecules that recruit circulating leukocytes and platelets to sites of vascular damage, tissue factor, which provides a procoagulant signal of vascular breach, and a surface upon which coagulation complexes are assembled. Blood flow is often characterized by 2 key variables: shear rate and shear stress. Shear rate affects several aspects of coagulation, including transport rates of platelets and plasma proteins to and from the injury site, platelet activation, and the kinetics of fibrin monomer formation and polymerization. Shear stress modulates adhesion rates of platelets and expression of adhesion molecules and procoagulant activity on endothelial cells lining the blood vessels. That no one abnormality in any component of Virchow's triad fully predicts coagulopathy a priori suggests coagulopathies are complex, multifactorial, and interactive. In this review, we focus on contributions of blood composition, vascular cells, and blood flow to hemostasis and thrombosis, and suggest that cross-talk among the 3 components of Virchow's triad is necessary for hemostasis and determines propensity for thrombosis or bleeding. Investigative models that permit interplay among these components are necessary to understand the operant pathophysiology, and effectively treat and prevent thrombotic and bleeding disorders.


Asunto(s)
Factores de Coagulación Sanguínea/metabolismo , Coagulación Sanguínea , Hemostasis , Trombosis/sangre , Plaquetas/metabolismo , Células Endoteliales/metabolismo , Hemodinámica , Humanos , Leucocitos/metabolismo , Trombosis/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...