Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Br J Haematol ; 173(5): 769-78, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27018425

RESUMEN

Haemostasis including blood coagulation is initiated upon vessel wall injury and indispensable to limit excessive blood loss. However, unregulated pathological coagulation may lead to vessel occlusion, causing thrombotic disorders, most notably myocardial infarction and stroke. Furthermore, blood exposure to foreign surfaces activates the intrinsic pathway of coagulation. Hence, various clinical scenarios, such as extracorporeal membrane oxygenation, require robust anticoagulation consequently leading to an increased bleeding risk. This study aimed to further assess the antithrombotic efficacy of the activated factor XII (FXIIa) inhibitor, rHA-Infestin-4, in several thrombosis models. In mice, rHA-Infestin-4 decreased occlusion rates in the mechanically-induced arterial (Folt's) and the FeCl3 -induced venous thrombosis model. rHA-Infestin-4 also protected from FeCl3 -induced arterial thrombosis and from stasis-prompted venous thrombosis in rabbits. Furthermore, rHA-Infestin-4 prevented occlusion in the arterio-venous shunt model in mice and rabbits where thrombosis was induced via a foreign surface. In contrast to heparin, the haemostatic capacity in rabbits was unaffected by rHA-Infestin-4. Using rodent and non-rodent species, our data demonstrate that the FXIIa inhibitor rHA-Infestin-4 decreased arterial, venous and foreign surface-induced thrombosis without affecting physiological haemostasis. Hence, we provide further evidence that targeting FXIIa represents a potent yet safe antithrombotic treatment approach, especially in foreign surface-triggered thrombosis.


Asunto(s)
Factor XIIa/antagonistas & inhibidores , Proteínas de Insectos/farmacología , Proteínas Recombinantes de Fusión/farmacología , Albúmina Sérica/farmacología , Trombosis/tratamiento farmacológico , Animales , Arteriopatías Oclusivas/tratamiento farmacológico , Arteriopatías Oclusivas/etiología , Modelos Animales de Enfermedad , Fibrinolíticos/farmacología , Hemostasis/efectos de los fármacos , Proteínas de Insectos/uso terapéutico , Cinética , Ratones , Conejos , Proteínas Recombinantes de Fusión/uso terapéutico , Albúmina Sérica/uso terapéutico , Albúmina Sérica Humana , Trombosis/etiología , Resultado del Tratamiento , Trombosis de la Vena/tratamiento farmacológico , Trombosis de la Vena/etiología
2.
Int J Med Microbiol ; 306(7): 572-579, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27338836

RESUMEN

Coagulation is a mechanism for wound healing after injury. Several recent studies delineate an additional role of the intrinsic pathway of coagulation, also known as the contact system, in the early innate immune response against bacterial infections. In this study, we investigated the role of factor XIII (FXIII), which is activated upon coagulation induction, during Streptococcus pyogenes-mediated skin and soft tissue infections. FXIII has previously been shown to be responsible for the immobilization of bacteria within a fibrin network which may prevent systemic bacterial dissemination. In order to investigate if the FXIII-mediated entrapment of S. pyogenes also influences the disease outcome we used a murine S. pyogenes M1 skin and soft tissue infection model. Here, we demonstrate that a lack of FXIII leads to prolonged clotting times, increased signs of inflammation, and elevated bacterial dissemination. Moreover, FXIII-deficient mice show an impaired survival when compared with wildtype animals. Additionally, local reconstitution of FXIII-deficient mice with a human FXIII-concentrate (Fibrogammin®P) could reduce the systemic complications, suggesting a protective role for FXIII during early S. pyogenes skin infection. FXIII therefore might be a possible therapeutically application to support the early innate immune response during skin infections caused by S. pyogenes.


Asunto(s)
Factor XIII/metabolismo , Interacciones Huésped-Patógeno , Inmunidad Innata , Enfermedades Cutáneas Bacterianas/inmunología , Infecciones Estreptocócicas/inmunología , Streptococcus pyogenes/inmunología , Animales , Antígenos Bacterianos , Proteínas de la Membrana Bacteriana Externa , Proteínas Portadoras , Modelos Animales de Enfermedad , Factor XIII/genética , Femenino , Ratones Endogámicos CBA , Ratones Noqueados , Enfermedades Cutáneas Bacterianas/microbiología , Enfermedades Cutáneas Bacterianas/patología , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/patología , Streptococcus pyogenes/patogenicidad , Análisis de Supervivencia
3.
Anesthesiology ; 122(2): 387-98, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25419685

RESUMEN

BACKGROUND: Edoxaban is an oral, selective direct factor Xa inhibitor approved in Japan for venous thromboembolism prevention after orthopedic surgery. Data are lacking regarding reversal strategies for edoxaban; this study assessed whether four-factor prothrombin complex concentrate (Beriplex/Kcentra; CSL Behring GmbH, Marburg, Germany) can effectively reverse its effects on hemostasis using a previously described rabbit model. METHODS: The study comprised assessments of thrombin generation in vitro, pharmacokinetic parameters, and edoxaban reversal in vivo. In a blinded in vivo stage, a standardized kidney incision was performed in animals (n = 11 per group) randomized to receive vehicle + saline, edoxaban (1,200 µg/kg) + saline, or edoxaban (1,200 µg/kg) + four-factor prothrombin complex concentrate (50 IU/kg). Animals were monitored for treatment impact on hemostasis and coagulation parameters. Data are median (range). Statistical tests were adjusted for multiple testing. RESULTS: Edoxaban administration increased blood loss (30 [2 to 44] ml) and time to hemostasis (23 [8.5 to 30.0] min) compared with the control group (3 [1 to 8] ml and 3 [2.0 to 5.0] min, respectively). Biomarkers of coagulation (prothrombin time, activated partial thromboplastin time, whole blood clotting time) and thrombin generation parameters (e.g., peak thrombin, endogenous thrombin potential, lag time) were also affected by edoxaban. Administration of four-factor prothrombin complex concentrate significantly reduced time to hemostasis (to 8 [6.5 to 14.0] min, observed P < 0.0001) and total blood loss (to 9 [4 to 22] ml, observed P = 0.0050) compared with the edoxaban + saline group. Of the biomarkers tested, prothrombin time, whole blood clotting time, and endogenous thrombin potential correlated best with clinical parameters. CONCLUSION: In a rabbit model of hemostasis, four-factor prothrombin complex concentrate administration significantly decreased edoxaban-associated hemorrhage.


Asunto(s)
Factores de Coagulación Sanguínea/uso terapéutico , Hemorragia/inducido químicamente , Hemorragia/prevención & control , Hemostáticos/uso terapéutico , Piridinas/antagonistas & inhibidores , Piridinas/toxicidad , Tiazoles/antagonistas & inhibidores , Tiazoles/toxicidad , Enfermedad Aguda , Animales , Factores de Coagulación Sanguínea/farmacocinética , Pruebas de Coagulación Sanguínea , Chinchilla , Determinación de Punto Final , Femenino , Trastornos Hemorrágicos/prevención & control , Hemostáticos/farmacocinética , Técnicas In Vitro , Piridinas/farmacocinética , Conejos , Tiazoles/farmacocinética , Trombina/metabolismo
4.
Circulation ; 127(20): 2038-46, 2013 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-23616627

RESUMEN

BACKGROUND: Exaggerated and prolonged inflammation after myocardial infarction (MI) accelerates left ventricular remodeling. Inflammatory pathways may present a therapeutic target to prevent post-MI heart failure. However, the appropriate magnitude and timing of interventions are largely unknown, in part because noninvasive monitoring tools are lacking. Here, we used nanoparticle-facilitated silencing of CCR2, the chemokine receptor that governs inflammatory Ly-6C(high) monocyte subset traffic, to reduce infarct inflammation in apolipoprotein E-deficient (apoE(-/-)) mice after MI. We used dual-target positron emission tomography/magnetic resonance imaging of transglutaminase factor XIII (FXIII) and myeloperoxidase (MPO) activity to monitor how monocyte subset-targeted RNAi altered infarct inflammation and healing. METHODS AND RESULTS: Flow cytometry, gene expression analysis, and histology revealed reduced monocyte numbers and enhanced resolution of inflammation in infarcted hearts of apoE(-/-) mice that were treated with nanoparticle-encapsulated siRNA. To follow extracellular matrix cross-linking noninvasively, we developed a fluorine-18-labeled positron emission tomography agent ((18)F-FXIII). Recruitment of MPO-rich inflammatory leukocytes was imaged with a molecular magnetic resonance imaging sensor of MPO activity (MPO-Gd). Positron emission tomography/magnetic resonance imaging detected anti-inflammatory effects of intravenous nanoparticle-facilitated siRNA therapy (75% decrease of MPO-Gd signal; P<0.05), whereas (18)F-FXIII positron emission tomography reflected unimpeded matrix cross-linking in the infarct. Silencing of CCR2 during the first week after MI improved ejection fraction on day 21 after MI from 29% to 35% (P<0.05). CONCLUSION: CCR2-targeted RNAi reduced recruitment of Ly-6C(high) monocytes, attenuated infarct inflammation, and curbed post-MI left ventricular remodeling.


Asunto(s)
Aterosclerosis/terapia , Marcación de Gen/métodos , Monocitos/metabolismo , Infarto del Miocardio/genética , Infarto del Miocardio/terapia , Interferencia de ARN/fisiología , Receptores CCR2/genética , Cicatrización de Heridas/genética , Secuencia de Aminoácidos , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Femenino , Predisposición Genética a la Enfermedad , Terapia Genética/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Monocitos/patología , Infarto del Miocardio/patología , Distribución Aleatoria , Receptores CCR2/antagonistas & inhibidores , Receptores CCR2/metabolismo
5.
Toxicol Appl Pharmacol ; 280(1): 70-7, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25102310

RESUMEN

Fibrinogen, a soluble 340kDa plasma glycoprotein, is critical in achieving and maintaining hemostasis. Reduced fibrinogen levels are associated with an increased risk of bleeding and recent research has investigated the efficacy of fibrinogen concentrate for controlling perioperative bleeding. European guidelines on the management of perioperative bleeding recommend the use of fibrinogen concentrate if significant bleeding is accompanied by plasma fibrinogen levels less than 1.5-2.0g/l. Plasma-derived human fibrinogen concentrate has been available for therapeutic use since 1956. The overall aim of the comprehensive series of non-clinical investigations presented was to evaluate i) the pharmacodynamic and pharmacokinetic characteristics and ii) the safety and tolerability profile of human fibrinogen concentrate Haemocomplettan P® (RiaSTAP®). Pharmacodynamic characteristics were assessed in rabbits, pharmacokinetic parameters were determined in rabbits and rats and a safety pharmacology study was performed in beagle dogs. Additional toxicology tests included: single-dose toxicity tests in mice and rats; local tolerance tests in rabbits; and neoantigenicity tests in rabbits and guinea pigs following the introduction of pasteurization in the manufacturing process. Human fibrinogen concentrate was shown to be pharmacodynamically active in rabbits and dogs and well tolerated, with no adverse events and no influence on circulation, respiration or hematological parameters in rabbits, mice, rats and dogs. In these non-clinical investigations, human fibrinogen concentrate showed a good safety profile. This data adds to the safety information available to date, strengthening the current body of knowledge regarding this hemostatic agent.


Asunto(s)
Fibrinógeno/metabolismo , Fibrinógeno/toxicidad , Animales , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Perros , Relación Dosis-Respuesta a Droga , Femenino , Fibrinógeno/farmacología , Cobayas , Hemostasis/efectos de los fármacos , Hemostasis/fisiología , Humanos , Masculino , Ratones , Conejos , Ratas , Ratas Wistar , Especificidad de la Especie
6.
Blood ; 118(9): 2589-98, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21613262

RESUMEN

Phylogenetically conserved serine protease cascades play an important role in invertebrate and vertebrate immunity. The mammalian coagulation system can be traced back some 400 million years and shares homology with ancestral serine proteinase cascades that are involved in, for example, Toll receptor signaling in insects and release of antimicrobial peptides during hemolymph clotting. In the present study, we show that the induction of coagulation by bacteria leads to immobilization and killing of Streptococcus pyogenes bacteria inside the clot. The entrapment is mediated via cross-linking of bacteria to fibrin fibers by the action of coagulation factor XIII (fXIII), an evolutionarily conserved transglutaminase. In a streptococcal skin infection model, fXIII(-/-) mice developed severe signs of pathologic inflammation at the local site of infection, and fXIII treatment of wild-type animals dampened bacterial dissemination during early infection. Bacterial killing and cross-linking to fibrin networks was also detected in tissue biopsies from patients with streptococcal necrotizing fasciitis, supporting the concept that coagulation is part of the early innate immune system.


Asunto(s)
Actividad Bactericida de la Sangre/inmunología , Coagulación Sanguínea/inmunología , Deficiencia del Factor XIII/inmunología , Factor XIII/fisiología , Fascitis Necrotizante/inmunología , Animales , Evolución Molecular , Deficiencia del Factor XIII/sangre , Fascitis Necrotizante/sangre , Fascitis Necrotizante/patología , Fascitis Necrotizante/terapia , Fibrina , Fibrinolisina/uso terapéutico , Humanos , Inflamación , Ratones , Ratones Endogámicos CBA , Ratones Noqueados , Filogenia , Especificidad de la Especie , Streptococcus pyogenes/inmunología , Trombina/farmacología
7.
Stroke ; 43(9): 2457-67, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22744646

RESUMEN

BACKGROUND AND PURPOSE: Inflammation and thrombosis are pathophysiological hallmarks of ischemic stroke still unamenable to therapeutic interventions. The contact-kinin system represents an interface between inflammatory and thrombotic circuits and is involved in stroke development. C1-inhibitor counteracts activation of the contact-kinin system at multiple levels. We investigated the therapeutic potential of C1-inhibitor in models of ischemic stroke. METHODS: Male and female C57Bl/6 mice and rats of different ages were subjected to middle cerebral artery occlusion and treated with C1-inhibitor after 1 hour or 6 hours. Infarct volumes and functional outcomes were assessed between day 1 and day 7, and findings were validated by magnetic resonance imaging. Blood-brain barrier damage, thrombus formation, and the local inflammatory response were determined poststroke. RESULTS: Treatment with 15.0 U C1-inhibitor, but not 7.5 U, 1 hour after stroke reduced infarct volumes by ≈60% and improved clinical scores in mice of either sex on day 1. This protective effect was preserved at later stages of infarction as well as in elderly mice and in another species, ie, rats. Delayed C1-inhibitor treatment still improved clinical outcome. Blood-brain barrier damage, edema formation, and inflammation were significantly lower compared with controls. Moreover, C1-inhibitor showed strong antithrombotic effects. CONCLUSIONS: C1-inhibitor is a multifaceted antiinflammatory and antithrombotic compound that protects from ischemic neurodegeneration in clinically meaningful settings.


Asunto(s)
Antiinflamatorios , Isquemia Encefálica/prevención & control , Proteína Inhibidora del Complemento C1/uso terapéutico , Fibrinolíticos , Daño por Reperfusión/prevención & control , Animales , Barrera Hematoencefálica/efectos de los fármacos , Western Blotting , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/patología , Isquemia Encefálica/patología , Femenino , Inmunohistoquímica , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/patología , Trombosis Intracraneal/tratamiento farmacológico , Trombosis Intracraneal/patología , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Examen Neurológico , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Daño por Reperfusión/patología , Caracteres Sexuales , Resultado del Tratamiento
8.
Am J Pathol ; 179(6): 3011-9, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22019897

RESUMEN

The transglutaminase-mediated, covalent cross-linking of proteins is an essential step in tissue remodeling after injury. This process provides tissues with extra rigidity and resistance against proteolytic degradation. Plasma coagulation factor XIII (FXIII) is a transglutaminase that promotes cross-linking of the extracellular matrix (ECM) components fibrin and fibronectin to form a provisional matrix in response to tissue damage. However, the functional requirement for this FXIII-mediated cross-linked provisional matrix in adult tissue remodeling remains to be defined. Although it has been proposed that the formation FXIII-mediated fibrin-fibronectin provisional matrix is a critical step for ECM remodeling, we show in an FXIII subunit A-deficient murine model of acute liver injury that the lack of FXIII subunit A did not interfere with collagen reconstruction and resolution after liver injury. Furthermore, FXIIIA deficiency caused significantly increased hepatocyte apoptosis and a delay in hepatocyte regeneration after injury, which were accompanied by a significantly high induction of p53 expression. These findings suggest novel functions of FXIII that the FXIII-mediated covalently cross-linked matrix could promote survival signals for hepatocytes in adult tissue remodeling.


Asunto(s)
Tetracloruro de Carbono/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/fisiopatología , Deficiencia del Factor XIII/fisiopatología , Factor XIII/fisiología , Animales , Apoptosis , Adhesión Celular/fisiología , Proliferación Celular , Transdiferenciación Celular/fisiología , Colágeno/fisiología , Reactivos de Enlaces Cruzados/metabolismo , Matriz Extracelular/fisiología , Factor XIIIa/fisiología , Fibrina/fisiología , Fibrinógeno/fisiología , Fibroblastos/fisiología , Fibronectinas/fisiología , Células Estrelladas Hepáticas/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Endogámicos CBA , Cicatrización de Heridas/fisiología
9.
Circulation ; 121(13): 1510-7, 2010 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-20308613

RESUMEN

BACKGROUND: Blood coagulation is a tightly regulated process of sequentially activated serine proteases culminating in fibrin formation, which is critical for limiting posttraumatic blood loss but also may contribute to acute thrombotic diseases, most notably myocardial infarction and stroke. Recent studies with factor XII-deficient mice revealed that the factor XII-induced intrinsic coagulation pathway is essential for pathological thrombus formation but dispensable for hemostasis. Consequently, these findings led to the hypothesis that factor XII could be a promising pharmacological target for safe antithrombotic therapy. METHODS AND RESULTS: The complementary DNA of the previously described factor XIIa inhibitor Infestin-4, cloned from the midgut of Triatoma infestans, was fused to recombinant human albumin (rHA) and analyzed in vitro. The resulting protein rHA-Infestin-4 specifically inhibits factor XIIa and causes prolonged activated partial thromboplastin time in human, mouse, and rat plasma. To assess its inhibitory potency in vivo, mice and rats were injected with rHA-Infestin-4 and challenged in pathological thrombus formation models. In addition, bleeding assays were performed. rHA-Infestin-4 completely abolished occlusive arterial thrombus formation in mice and rats while leaving hemostasis fully intact. Furthermore, rHA-Infestin-4 was highly protective in a murine model of ischemic stroke. CONCLUSIONS: These results identify rHA-Infestin-4 as a promising agent to achieve powerful protection from ischemic cardiovascular and cerebrovascular events without affecting hemostasis.


Asunto(s)
Albúminas/farmacología , Arteriopatías Oclusivas/tratamiento farmacológico , Factor XIIa/antagonistas & inhibidores , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Proteínas de Insectos/farmacología , Trombosis/tratamiento farmacológico , Albúminas/genética , Animales , Arteriopatías Oclusivas/sangre , Isquemia Encefálica/sangre , Isquemia Encefálica/tratamiento farmacológico , ADN Complementario , Factor XIIa/genética , Factor XIIa/metabolismo , Hemostasis/efectos de los fármacos , Humanos , Infarto de la Arteria Cerebral Media/sangre , Proteínas de Insectos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Tiempo de Tromboplastina Parcial , Ratas , Ratas Endogámicas , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Trombosis/sangre
10.
PLoS One ; 16(10): e0258192, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34614035

RESUMEN

OBJECTIVES: Acquired coagulopathy may be associated with bleeding risk. Approaches to restore haemostasis include administration of coagulation factor concentrates, but there are concerns regarding potential prothrombotic risk. The present study assessed the prothrombotic potential of four-factor prothrombin complex concentrate (4F-PCC) versus activated PCC (aPCC) and recombinant factor VIIa (rFVIIa), using three preclinical animal models. METHODS: The first model was a modified Wessler model of venous stasis-induced thrombosis in rabbit, focusing on dilutional coagulopathy; the second model employed the same system but focused on direct oral anticoagulant reversal (i.e. edoxaban). The third model assessed the prothrombotic impact of 4F-PCC, aPCC and rFVIIa in a rat model of ferric chloride-induced arterial thrombosis. RESULTS: In the first model, thrombi were observed at aPCC doses ≥10 IU/kg (therapeutic dose 100 IU/kg) and rFVIIa doses ≥50 µg/kg (therapeutic dose 90 µg/kg), but not 4F-PCC 50 IU/kg (therapeutic dose 50 IU/kg). The impact of 4F-PCC (up to 300 IU/kg) on thrombus formation was evident from 10 minutes post-administration, but not at 24 hours post-administration; this did not change with addition of tranexamic acid and/or fibrinogen concentrate. 4F-PCC-induced thrombus formation was lower after haemodilution versus non-haemodilution. In the second model, no prothrombotic effect was confirmed at 4F-PCC 50 IU/kg. The third model showed lower incidence of thrombus formation for 4F-PCC 50 IU/kg versus aPCC (50 U/kg) and rFVIIa (90 µg/kg). CONCLUSIONS: These results suggest that 4F-PCC has a low thrombotic potential versus aPCC or rFVIIa, supporting the clinical use of 4F-PCC for the treatment of coagulopathy-mediated bleeding.


Asunto(s)
Factores de Coagulación Sanguínea/metabolismo , Trombosis/metabolismo , Animales , Arterias/efectos de los fármacos , Arterias/patología , Factor VIIa/farmacología , Femenino , Hemodilución , Conejos , Ratas Sprague-Dawley , Proteínas Recombinantes/farmacología , Trombosis/patología , Factores de Tiempo , Ácido Tranexámico/farmacología
11.
J Trauma ; 68(5): 1151-7, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-19996804

RESUMEN

BACKGROUND: Fluid resuscitation after traumatic injury may necessitate coagulation factor replacement to prevent bleeding complications of dilutional coagulopathy. Recombinant activated factor VII (rFVIIa) is being widely investigated as a hemostatic agent in trauma. Multicomponent therapy with prothrombin complex concentrate (PCC) containing coagulation factors II, VII, IX, and X might offer potential advantages. METHODS: Anesthetized mildly hypothermic normotensive pigs were hemodiluted by substituting 65% to 70% of total blood volume in phases with hydroxyethyl starch and red cells. Thereafter, animals received 12.5 mL . kg isotonic saline placebo, 35 IU . kg PCC, or 180 microg x kg rFVIIa. Immediately afterward, a standardized spleen injury was inflicted, and prothrombin time (PT) and hemostasis were assessed. Thrombin generation was also determined. RESULTS: Hemodilution depleted levels of factors II, VII, IX, and X markedly, prolonged PT and decreased thrombin formation. PCC and rFVIIa both fully normalized the hemodilution-induced lengthening of PT. In PCC recipients, peak thrombin generation was greater by a median of 60.7 nM (confidence interval 56.4-64.9 nM) compared with the rFVIIa group (p = 0.008). After spleen trauma, time to hemostasis was shortened to a median of 35 minutes in animals treated with PCC versus 94 minutes with rFVIIa (p = 0.016). CONCLUSIONS: In a pilot study involving an in vivo large-animal model of spleen trauma, PCC accelerated hemostasis and augmented thrombin generation compared with rFVIIa. Further investigations are warranted on PCC as a hemostatic agent in trauma.


Asunto(s)
Trastornos de la Coagulación Sanguínea/tratamiento farmacológico , Factores de Coagulación Sanguínea/uso terapéutico , Modelos Animales de Enfermedad , Factor VIIa/uso terapéutico , Hemodilución/efectos adversos , Heridas y Lesiones/complicaciones , Animales , Trastornos de la Coagulación Sanguínea/sangre , Trastornos de la Coagulación Sanguínea/etiología , Evaluación Preclínica de Medicamentos , Fluidoterapia/efectos adversos , Fluidoterapia/métodos , Hemodilución/métodos , Hemorragia/complicaciones , Hemorragia/terapia , Hemostasis/efectos de los fármacos , Estimación de Kaplan-Meier , Masculino , Proyectos Piloto , Tiempo de Protrombina , Proteínas Recombinantes/uso terapéutico , Resucitación/efectos adversos , Resucitación/métodos , Bazo/lesiones , Estadísticas no Paramétricas , Porcinos , Factores de Tiempo , Resultado del Tratamiento , Heridas y Lesiones/terapia
12.
J Pharmacol Toxicol Methods ; 105: 106901, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32771565

RESUMEN

INTRODUCTION: The implementation of novel, reliable biomarkers for the early and differential diagnosis of acute kidney injury (AKI) could greatly improve the timely treatment and prevention of disease progression, particularly since the current gold standards for detecting kidney injury such as serum creatinine (SCr) and blood urea nitrogen (BUN) lack sensitivity and specificity. We evaluated novel urinary kidney injury biomarkers focusing on early detection and better prediction of AKI with higher sensitivity and specificity. METHODS: In the rat, urinary biomarkers for kidney injury, i.e. albumin, beta-2-microglobulin (B2M), clusterin, cystatin C, kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), osteopontin (OPN), and total protein (TP), were investigated in an AKI model using different hyperosmolar and high-dose solutions, i.e. mannitol, sucrose, and contrast medium (CM), as acute single insults leading to kidney injury. Additionally, dose-dependency of sucrose was investigated and effects were compared to the sucrose- and iron-containing marketed drug Venofer®. RESULTS: Levels of excreted urinary biomarkers correlated with severity of AKI, exhibited a dose-dependent response to sucrose treatment, and demonstrated evidence of recovery from kidney injury with transient and reversible changes. The exceptions were KIM-1 and NGAL, which showed later responses following CM and iron-induced renal injury. All biomarkers outperformed plasma creatinine (PCr), BUN, and histopathology, with regard to practicability and/or detection of proximal tubular injury. DISCUSSION: The use of a panel of urinary kidney injury biomarkers emerged as an early, sensitive, and predictive tool to detect AKI showing enhanced sensitivity compared to current state-of-the-art markers.


Asunto(s)
Lesión Renal Aguda/sangre , Lesión Renal Aguda/metabolismo , Biomarcadores/sangre , Biomarcadores/metabolismo , Riñón/metabolismo , Animales , Nitrógeno de la Urea Sanguínea , Moléculas de Adhesión Celular/metabolismo , Creatinina/sangre , Modelos Animales de Enfermedad , Diagnóstico Precoz , Pruebas de Función Renal/métodos , Lipocalina 2/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Sensibilidad y Especificidad
13.
J Thromb Haemost ; 18(12): 3194-3202, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32810892

RESUMEN

BACKGROUND: Hemophilia B is caused by coagulation factor IX (FIX) deficiency. Recombinant fusion protein linking coagulation FIX with recombinant albumin (rIX-FP; Idelvion® ) is used for replacement therapy with an extended half-life. A previous quantitative whole-body autoradiography (QWBA) study investigating the biodistribution of rIX-FP indicated equal biodistribution, but more prolonged tissue retention compared with a marketed recombinant FIX product. OBJECTIVES: To complete and confirm the QWBA study data by directly measuring rIX-FP protein and activity levels in tissues following intravenous (i.v.) administration to normal rats and FIX-deficient (hemophilia B) mice. METHODS: After i.v. administration of rIX-FP at a dose of 2000 IU/kg, animals were euthanized at specific time points up to 72 hours postdosing. Subsequently, plasma and various tissues, which were selected based on the previous QWBA results, were harvested and analyzed for FIX antigen levels using an ELISA (both species) or an immunohistochemistry method (mice only), as well as for FIX activity levels (mice only) using a chromogenic assay. RESULTS: In rats, rIX-FP distributed extravascularly into all tissues analyzed (ie, liver, kidney, skin and knee) with peak antigen levels reached between 1 and 7 hours postdosing. In hemophilia B mice, rIX-FP tissue distribution was comparable to rats. FIX antigen levels correlated well with FIX activity readouts. CONCLUSIONS: Our results confirm QWBA data showing that rIX-FP distributes into relevant target tissues. Importantly, it was demonstrated that rIX-FP available in tissues retains its functional activity and can thus facilitate its therapeutic activity at sites of potential injury.


Asunto(s)
Hemofilia B , Roedores , Administración Intravenosa , Animales , Factor IX/metabolismo , Semivida , Hemofilia B/tratamiento farmacológico , Ratones , Ratas , Proteínas Recombinantes de Fusión/uso terapéutico , Roedores/metabolismo , Distribución Tisular
14.
Thromb Res ; 122(1): 117-23, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-17977586

RESUMEN

INTRODUCTION: Warfarin and its derivatives are widely used for prevention of thrombotic incidents. Prothrombin complex concentrate (PCC) and recombinant activated factor VII (rFVIIa) have been used clinically for the acute reversal of this agent but there is a paucity of data on comparative efficacies of these hemostatic interventions. MATERIALS AND METHODS: Using in vivo rat and in vitro human models of anticoagulation, we compared PCC and rFVIIa on the recovery of endogenous thrombin generation. For in vivo reversal of anticoagulation, saline (control), PCC 50 U ml(-1), or rFVIIa100 mug ml(-1) was given to rats which received phenprocoumon (2.5 mg kg(-1)) orally. For in vitro model, plasma samples from warfarin-treated individuals with INR values of 2.1-6.7 were spiked with PCC (0.2, 0.4, or 0.72 U ml(-1)) or rFVIIa (3.0 mug/ml). The treatments were evaluated using prothrombin time (PT) and thrombin generation (Thrombinoscope). RESULTS: Administration of phenprocoumon to rats prolonged PT (14.7+/-0.5 to 50.43+/-0.7 s) and decreased peak thrombin generation by 89+/-2.3%. Administration of PCC dose dependently reversed the anticoagulation effects both in warfarin-treated human plasma and in phenprocoumon-treated rats by shortening PT and increasing peak thrombin levels. However, rFVIIa only reversed PT, but had minimal effects on peak thrombin levels. CONCLUSION: Both PCC and rFVIIa reverse warfarin anticoagulation based on PT, but only PCC restores overall thrombin generation.


Asunto(s)
Factor VIIa/metabolismo , Protrombina/fisiología , Vitamina K/antagonistas & inhibidores , Animales , Anticoagulantes/uso terapéutico , Factor VIIa/efectos de los fármacos , Humanos , Relación Normalizada Internacional , Modelos Animales , Protrombina/efectos de los fármacos , Tiempo de Protrombina , Ratas , Proteínas Recombinantes/metabolismo , Trombina/efectos de los fármacos , Trombina/metabolismo , Warfarina/uso terapéutico
15.
Anesth Analg ; 106(4): 1070-7, table of contents, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18349175

RESUMEN

BACKGROUND: In this study, we used a porcine model to investigate whether impaired coagulation and severe arterial or venous bleeding could be normalized by substitution with a prothrombin complex concentrate (PCC), Beriplex P/N, containing coagulation factors II, VII, IX, and X. METHODS: Dilutional coagulopathy was induced in anesthetized pigs by fractionated blood withdrawal (approximately 65% of total volume), followed by erythrocyte retransfusion and volume substitution with a total of 1000 mL of hydroxyethyl starch (Infukoll 6%). Animals were randomized to no treatment, treatment with placebo, or treatment with 35 U/kg PCC. Arterial (spleen incision) or venous (bone injury) bleeding was inflicted. Thromboelastometry, hematology, and coagulation tests were performed at baseline, after dilution, and after study treatments had been administered and injury inflicted. The primary end-point was postinjury time to hemostasis. RESULTS: Hemodilution resulted in a decrease in coagulation factor concentrations to approximately 35% and prolonged prothrombin time. Platelet numbers decreased from approximately 400,000 to approximately 100,000/microL, and aggregation and adhesion were impaired. PCC effectively substituted the deficient prothrombin factors (II, VII, IX, and X) and normalized the prolonged prothrombin time. After spleen injury, PCC significantly reduced time to hemostasis versus dilutional control (median, 35 vs 82.5 min; P < 0.0001), and produced a nonsignificant trend towards reduction in blood loss (mean, 275 vs 589 mL). PCC also significantly reduced time to hemostasis (median, 27 vs 97 min; P < 0.0011) and blood loss (mean, 71 vs 589 mL; P < 0.0017) after bone injury. CONCLUSIONS: Dilutional coagulopathy produced a generalized decrease in coagulation factors and impaired platelet function. Substitution with PCC effectively normalized coagulation and significantly improved hemostasis after venous and arterial bleeding.


Asunto(s)
Trastornos de la Coagulación Sanguínea/tratamiento farmacológico , Factores de Coagulación Sanguínea/uso terapéutico , Anestésicos Intravenosos , Animales , Modelos Animales de Enfermedad , Hemodilución/métodos , Hemorragia/fisiopatología , Agregación Plaquetaria/efectos de los fármacos , Porcinos , Tiopental
16.
Circulation ; 113(9): 1196-202, 2006 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-16505171

RESUMEN

BACKGROUND: Identification of key molecular players in myocardial healing could lead to improved therapies, reduction of scar formation, and heart failure after myocardial infarction (MI). We hypothesized that clotting factor XIII (FXIII), a transglutaminase involved in wound healing, may play an important role in MI given prior clinical and mouse model data. METHODS AND RESULTS: To determine whether a truly causative relationship existed between FXIII activity and myocardial healing, we prospectively studied myocardial repair in FXIII-deficient mice. All FXIII(-/-) and FXIII(-)(/+) (FXIII activity <5% and 70%) mice died within 5 days after MI from left ventricular rupture. In contradistinction, FXIII(-/-) mice that received 5 days of intravenous FXIII replacement therapy had normal survival rates; however, cardiac MRI demonstrated worse left ventricular remodeling in these reconstituted FXIII(-/-) mice. Using a FXIII-sensitive molecular imaging agent, we found significantly greater FXIII activity in wild-type mice and FXIII(-/-) mice receiving supplemental FXIII than in FXIII(-/-) mice (P<0.05). In FXIII(-/-) but not in reconstituted FXIII(-/-) mice, histology revealed diminished neutrophil migration into the MI. Reverse transcriptase-polymerase chain reaction studies suggested that the impaired inflammatory response in FXIII(-/-) mice was independent of intercellular adhesion molecule and lipopolysaccharide-induced CXC chemokine, both important for cell migration. After MI, expression of matrix metalloproteinase-9 was 650% higher and collagen-1 was 53% lower in FXIII(-/-) mice, establishing an imbalance in extracellular matrix turnover and providing a possible mechanism for the observed cardiac rupture in the FXIII(-/-) mice. CONCLUSIONS: These data suggest that FXIII has an important role in murine myocardial healing after infarction.


Asunto(s)
Deficiencia del Factor XIII/complicaciones , Rotura Cardíaca Posinfarto/etiología , Infarto del Miocardio/complicaciones , Remodelación Ventricular , Cicatrización de Heridas , Animales , Quimiotaxis de Leucocito , Proteínas de la Matriz Extracelular/análisis , Proteínas de la Matriz Extracelular/genética , Factor XIII/fisiología , Ratones , Ratones Noqueados , Neutrófilos/fisiología
17.
Thromb Res ; 119(5): 643-51, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-16842841

RESUMEN

INTRODUCTION: Prothrombin complex concentrate (PCC) is recommended for emergency reversal of oral coumarin anticoagulation. Recently, recombinant factor VIIa (rFVIIa) has also been investigated for this purpose, although no direct comparison of PCC and rFVIIa has been reported. This study was designed to compare the effectiveness of PCC and rFVIIa for reversal of both acute and sustained coumarin anticoagulation. MATERIALS AND METHODS: In the acute model, rats received 2.5 mg.kg(-1) phenprocoumon, and reversal of anticoagulation by 4.88 mL.kg(-1) saline, 100 microg.kg(-1) rFVIIa (NovoSeven) or 50 U.kg(-1) PCC (Beriplex P/N) was assessed at 16 h. For the sustained model, a second phenprocoumon dose was administered at 24 h and anticoagulation reversal evaluated at 48 h. Study endpoints were activated partial thromboplastin time (aPTT), prothrombin time (PT) and tail tip bleeding. RESULTS: Acute anticoagulation raised median PT to 4.3 fold the normal level. This elevation was nearly completely reversed both by rFVIIa and PCC. aPTT increase was minor. Effects of sustained anticoagulation were more severe and pervasive, with aPTT, PT and blood loss increasing to 7.7, 51 and 30 fold the control levels, respectively. In the sustained model, rFVIIa substantially reduced and PCC fully normalized PT. In this model, PCC also diminished aPTT (p<0.01), fully normalized blood loss (p<0.01) and shortened bleeding time (p=0.008), while rFVIIa was without significant effect on these endpoints. CONCLUSIONS: In a sustained anticoagulation animal model designed to simulate standard long-term oral coumarin therapy in patients, PCC was more effective than rFVIIa in restoring hemostatic function.


Asunto(s)
Anticoagulantes/antagonistas & inhibidores , Factores de Coagulación Sanguínea/administración & dosificación , Factor VII/administración & dosificación , Fenprocumón/antagonistas & inhibidores , Animales , Anticoagulantes/administración & dosificación , Tiempo de Sangría , Modelos Animales de Enfermedad , Esquema de Medicación , Factor VIIa , Femenino , Estimación de Kaplan-Meier , Tiempo de Tromboplastina Parcial , Fenprocumón/administración & dosificación , Tiempo de Protrombina , Ratas , Proteínas Recombinantes/administración & dosificación , Reproducibilidad de los Resultados , Tasa de Supervivencia
18.
Thromb Haemost ; 95(3): 546-50, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16525585

RESUMEN

Thrombin-activated Factor XIII (FXIIIa), a plasma transglutaminase, stabilizes fibrin clots by crosslinking fibrin chains. FXIIIa was previously shown by us to exhibit proangiogenic activity associated with downregulation of thrombospondin-1, phosphorylation of vascular endothelial growth factor receptor 2 (VEGFR-2), and upregulation of c-Jun. In the current study, we evaluated the proangiogenic effect of FXIIIa in two murine models: a neonatal heterotopic cardiac allograft model in normal mice, and a Matrigel plug model in FXIII-deficient mice. In the neonatal cardiac allograft model, the number of new vessels as well as graft viability (contractile performance) was significantly higher in FXIIIa-injected animals than in controls. A significant increase in the level of c-Jun mRNA and a significant decrease in the level of TSP-1 mRNA were observed in heart allografts treated with FXIIIa. A marked decrease in TSP-1 protein expression was observed within the endothelial cells of hearts treated with FXIIIa. In the Matrigel plug model, FXIII-deficient mice showed a significantly decreased number of new vessels compared to that of the control mice, and the number of vessels almost reached normal levels following addition of FXIIIa. The results of this study provide substantial in vivo evidence for the proangiogenic activity of FXIIIa.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Deficiencia del Factor XIII/genética , Factor XIIIa/farmacología , Trasplante de Corazón , Neovascularización Fisiológica/efectos de los fármacos , Trasplante Heterotópico , Animales , Animales Recién Nacidos , Colágeno , Vasos Coronarios/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Oído Externo , Factor XIII/genética , Supervivencia de Injerto/efectos de los fármacos , Laminina , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Modelos Animales , Miocardio/metabolismo , Proteoglicanos , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , ARN Mensajero/metabolismo , Trombospondina 1/genética , Trombospondina 1/metabolismo
19.
Pharmacol Res Perspect ; 4(2): e00227, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27069637

RESUMEN

Factor XIII (FXIII) is a coagulation protein which plays a major role in hemostasis by covalently cross-linking fibrin molecules, thereby stabilizing the blood clot and increasing resistance to fibrinolysis. FXIII deficiency, either congenital or acquired, is associated with spontaneous bleeding, increased bleeding time, and poor wound healing. Purified plasma-derived human FXIII concentrate (pd hFXIII) has been available since 1993 for therapeutic use in congenital FXIII deficiency. This set of nonclinical investigations aimed to evaluate the pharmacodynamic effects and assess the safety profile of pd hFXIII. The efficacy and safety of pd hFXIII were evaluated by pharmacodynamic, pharmacokinetic, and toxicity studies in mice and rats, safety pharmacology studies in dogs, neoantigenicity study, local tolerance, and thrombogenicity tests in rabbits. Administration of pd hFXIII resulted in the correction of deficits in clot formation kinetics and strength as measured by thromboelastometry, and was not associated with thrombus formation up to 350 IU/kg in FXIII knockout mice. There was no production of neoantigens resulting from the viral elimination manufacturing steps detected, and no adverse reactions were observed in toxicity studies with single doses up to 3550 IU/kg in mice and 1420 IU/kg in rats; nor from repeat doses of 350 IU/kg in rats. In addition, local tolerance tests revealed a good tolerability profile in rabbits. Overall, this data showed that pd hFXIII was well tolerated and pharmacodynamically active in preclinical animal models, supporting pd hFXIII as a therapy for FXIII deficiency.

20.
PLoS One ; 11(1): e0146783, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26815580

RESUMEN

BACKGROUND AND PURPOSE: Ischemic stroke provokes severe brain damage and remains a predominant disease in industrialized countries. The coagulation factor XII (FXII)-driven contact activation system plays a central, but not yet fully defined pathogenic role in stroke development. Here, we investigated the efficacy of the FXIIa inhibitor rHA-Infestin-4 in a rat model of ischemic stroke using both a prophylactic and a therapeutic approach. METHODS: For prophylactic treatment, animals were treated intravenously with 100 mg/kg rHA-Infestin-4 or an equal volume of saline 15 min prior to transient middle cerebral artery occlusion (tMCAO) of 90 min. For therapeutic treatment, 100 mg/kg rHA-Infestin-4, or an equal volume of saline, was administered directly after the start of reperfusion. At 24 h after tMCAO, rats were tested for neurological deficits and blood was drawn for coagulation assays. Finally, brains were removed and analyzed for infarct area and edema formation. RESULTS: Within prophylactic rHA-Infestin-4 treatment, infarct areas and brain edema formation were reduced accompanied by better neurological scores and survival compared to controls. Following therapeutic treatment, neurological outcome and survival were still improved although overall effects were less pronounced compared to prophylaxis. CONCLUSIONS: With regard to the central role of the FXII-driven contact activation system in ischemic stroke, inhibition of FXIIa may represent a new and promising treatment approach to prevent cerebral ischemia/reperfusion injury.


Asunto(s)
Factor XIIa/antagonistas & inhibidores , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Proteínas de Insectos/farmacología , Proteínas Recombinantes de Fusión/farmacología , Daño por Reperfusión/prevención & control , Inhibidores de Serina Proteinasa/farmacología , Albúmina Sérica/farmacología , Animales , Encéfalo/irrigación sanguínea , Encéfalo/efectos de los fármacos , Encéfalo/patología , Células CHO , Cricetulus , Evaluación Preclínica de Medicamentos , Factor XIIa/metabolismo , Proteínas de Insectos/uso terapéutico , Masculino , Ratas , Proteínas Recombinantes de Fusión/uso terapéutico , Prueba de Desempeño de Rotación con Aceleración Constante , Inhibidores de Serina Proteinasa/uso terapéutico , Albúmina Sérica/uso terapéutico , Albúmina Sérica Humana
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA