Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(5)2023 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-36902231

RESUMEN

Macrophages express the A subunit of coagulation factor XIII (FXIII-A), a transglutaminase which cross-links proteins through Nε-(γ-L-glutamyl)-L-lysyl iso-peptide bonds. Macrophages are major cellular constituents of the atherosclerotic plaque; they may stabilize the plaque by cross-linking structural proteins and they may become transformed into foam cells by accumulating oxidized LDL (oxLDL). The combination of oxLDL staining by Oil Red O and immunofluorescent staining for FXIII-A demonstrated that FXIII-A is retained during the transformation of cultured human macrophages into foam cells. ELISA and Western blotting techniques revealed that the transformation of macrophages into foam cells elevated the intracellular FXIII-A content. This phenomenon seems specific for macrophage-derived foam cells; the transformation of vascular smooth muscle cells into foam cells fails to induce a similar effect. FXIII-A containing macrophages are abundant in the atherosclerotic plaque and FXIII-A is also present in the extracellular compartment. The protein cross-linking activity of FXIII-A in the plaque was demonstrated using an antibody labeling the iso-peptide bonds. Cells showing combined staining for FXIII-A and oxLDL in tissue sections demonstrated that FXIII-A-containing macrophages within the atherosclerotic plaque are also transformed into foam cells. Such cells may contribute to the formation of lipid core and the plaque structurization.


Asunto(s)
Aterosclerosis , Factor XIII , Placa Aterosclerótica , Humanos , Aterosclerosis/metabolismo , Factor XIII/metabolismo , Células Espumosas/metabolismo , Lipoproteínas LDL/metabolismo , Macrófagos/metabolismo , Péptidos/metabolismo , Placa Aterosclerótica/metabolismo
2.
Int J Mol Sci ; 23(10)2022 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-35628664

RESUMEN

Plasma factor XIII (pFXIII) is a heterotetramer of FXIII-A and FXIII-B subunits. The cellular form (cFXIII), a dimer of FXIII-A, is present in a number of cell types. Activated FXIII (FXIIIa), a transglutaminase, plays an important role in clot stabilization, wound healing, angiogenesis and maintenance of pregnancy. It has a direct effect on vascular endothelial cells and fibroblasts, which have been implicated in the development of atherosclerotic plaques. Our aim was to explore the effect of FXIIIa on human aortic smooth muscle cells (HAoSMCs), another major cell type in the atherosclerotic plaque. Osteoblastic transformation induced by Pi and Ca2+ failed to elicit the expression of cFXIII in HAoSMCs. EZ4U, CCK-8 and CytoSelect Wound Healing assays were used to investigate cell proliferation and migration. The Sircol Collagen Assay Kit was used to monitor collagen secretion. Thrombospondin-1 (TSP-1) levels were measured by ELISA. Cell-associated TSP-1 was detected by the immunofluorescence technique. The TSP-1 mRNA level was estimated by RT-qPCR. Activated recombinant cFXIII (rFXIIIa) increased cell proliferation and collagen secretion. In parallel, a 67% decrease in TSP-1 concentration in the medium and a 2.5-fold increase in cells were observed. TSP-1 mRNA did not change significantly. These effects of FXIIIa might contribute to the pathogenesis of atherosclerotic plaques.


Asunto(s)
Factor XIIIa , Placa Aterosclerótica , Transglutaminasas , Colágeno , Células Endoteliales/metabolismo , Factor XIIIa/genética , Factor XIIIa/metabolismo , Humanos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , ARN Mensajero/metabolismo , Trombospondina 1/genética , Transglutaminasas/genética , Transglutaminasas/metabolismo
3.
Int J Mol Sci ; 21(22)2020 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-33182600

RESUMEN

Hemostasis disorder in patients with end-stage renal disease (ESRD) is frequently associated with bleeding diathesis but it may also manifest in thrombotic complications. Analysis of individual coagulation and fibrinolytic factors may shed light on the background of this paradox situation. Here we explored components essential for fibrin formation/stabilization in ESRD patients being on maintenance hemodiafiltration (HDF) or hemodialysis (HD). Pre-dialysis fibrinogen, factor XIII (FXIII) antigen concentrations and FXIII activity were elevated, while α2-plasmin inhibitor (α2PI) activity decreased. The inflammatory status, as characterized by C-reactive protein (CRP) was a key determinant of fibrinogen concentration, but not of FXIII and α2PI levels. During a 4-h course of HDF or HD, fibrinogen concentration and FXIII levels gradually elevated. When compensated for the change in plasma water, i.e., normalized for plasma albumin concentration, only FXIII elevation remained significant. There was no difference between HDF and HD treatments. Individual HDF treatment did not influence α2PI activity, however after normalization it decreased significantly. HD treatment had a different effect, α2PI activities became elevated but the elevation disappeared after normalization. Elevated fibrinogen and FXIII levels in ESRD patients might contribute to the increased thrombosis risk, while decreased α2PI activity might be associated with elevated fibrinolytic potential.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Fallo Renal Crónico/sangre , Fallo Renal Crónico/terapia , Adolescente , Adulto , Anciano , Coagulación Sanguínea , Proteína C-Reactiva/metabolismo , Factor XIII/metabolismo , Femenino , Fibrinógeno/metabolismo , Fibrinólisis , Hemodiafiltración , Hemorragia/sangre , Hemorragia/etiología , Humanos , Fallo Renal Crónico/congénito , Masculino , Persona de Mediana Edad , Diálisis Renal , Factores de Riesgo , Trombosis/sangre , Trombosis/etiología , Adulto Joven , alfa 2-Antiplasmina/metabolismo
4.
Int J Mol Sci ; 20(23)2019 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-31783511

RESUMEN

Cellular factor XIII (cFXIII, FXIII-A2), a transglutaminase, has been demonstrated in a few cell types. Its main function is to cross-link proteins by isopeptide bonds. Here, we investigated the presence of cFXIII in cells of human cornea. Tissue sections of the cornea were immunostained for FXIII-A in combination with staining for CD34 antigen or isopeptide cross-links. Isolated corneal keratocytes were also evaluated by immunofluorescent microscopy and flow cytometry. FXIII-A in the corneal stroma was quantified by Western blotting. FXIII-A mRNA was detected by RT-qPCR. The cornea of FXIII-A-deficient patients was evaluated by cornea topography. FXIII-A was detected in 68 ± 13% of CD34+ keratocytes. Their distribution in the corneal stroma was unequal; they were most abundant in the subepithelial tertile. cFXIII was of cytoplasmic localization. In the stroma, 3.64 ng cFXIII/mg protein was measured. The synthesis of cFXIII by keratocytes was confirmed by RT-qPCR. Isopeptide cross-links were detected above, but not within the corneal stroma. Slight abnormality of the cornea was detected in six out of nine FXIII-A-deficient patients. The presence of cFXIII in human keratocytes was established for the first time. cFXIII might be involved in maintaining the stability of the cornea and in the corneal wound healing process.


Asunto(s)
Queratocitos de la Córnea/metabolismo , Sustancia Propia/metabolismo , Factor XIII/metabolismo , Transglutaminasas/metabolismo , Pruebas de Coagulación Sanguínea/métodos , Lesiones de la Cornea/metabolismo , Humanos , ARN Mensajero/metabolismo , Cicatrización de Heridas/fisiología
5.
Physiol Rev ; 91(3): 931-72, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21742792

RESUMEN

Factor XIII (FXIII) is unique among clotting factors for a number of reasons: 1) it is a protransglutaminase, which becomes activated in the last stage of coagulation; 2) it works on an insoluble substrate; 3) its potentially active subunit is also present in the cytoplasm of platelets, monocytes, monocyte-derived macrophages, dendritic cells, chondrocytes, osteoblasts, and osteocytes; and 4) in addition to its contribution to hemostasis, it has multiple extra- and intracellular functions. This review gives a general overview on the structure and activation of FXIII as well as on the biochemical function and downregulation of activated FXIII with emphasis on new developments in the last decade. New aspects of the traditional functions of FXIII, stabilization of fibrin clot, and protection of fibrin against fibrinolysis are summarized. The role of FXIII in maintaining pregnancy, its contribution to the wound healing process, and its proangiogenic function are reviewed in details. Special attention is given to new, less explored, but promising fields of FXIII research that include inhibition of vascular permeability, cardioprotection, and its role in cartilage and bone development. FXIII is also considered as an intracellular enzyme; a separate section is devoted to its intracellular activation, intracellular action, and involvement in platelet, monocyte/macrophage, and dendritic cell functions.


Asunto(s)
Fenómenos Fisiológicos Celulares , Factor XIII/metabolismo , Plasma/fisiología , Animales , Células Sanguíneas/fisiología , Coagulación Sanguínea/fisiología , Desarrollo Óseo/fisiología , Permeabilidad Capilar/fisiología , Cardiotónicos/metabolismo , Cartílago/crecimiento & desarrollo , Regulación hacia Abajo , Factor XIII/química , Factor XIIIa/metabolismo , Femenino , Fibrina/metabolismo , Humanos , Neovascularización Fisiológica , Embarazo/fisiología , Cicatrización de Heridas/fisiología
6.
Mol Cell Biochem ; 448(1-2): 199-209, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29484525

RESUMEN

Factor XIII (FXIII) stabilizes and protects the fibrin network. Its role in myocardial infarction (MI) is still to be clarified. To evaluate the association of FXIII levels with MI in young patients and to investigate how the FXIII-A p.Val34Leu, FXIII-B p.His95Arg, and IVS11, c.1952 + 144 C>G (Intron K) polymorphisms influence FXIII levels and MI risk. Patients with ST elevation MI below 40 years of age (MI, n = 119), age-matched clinical controls (CC, n = 101) without MI and coronary artery disease, and healthy controls (HC, n = 120) were investigated for FXIII activity, FXIII-A2B2, FXIII-B concentrations and for the polymorphisms. FXIII activity and FXIII-A2B2 antigen were significantly elevated in MI. FXIII activity and antigen were significantly elevated in Arg95, while decreased in Intron K "G" carriers. Smoking had an independent increasing effect on FXIII activity and FXIII-A2B2 antigen. Intron K C>G polymorphism significantly decreased the risk of MI in patients with elevated fibrinogen. Among the investigated factors Intron K C>G polymorphism and smoking have the most powerful effect on FXIII levels and on the risk of MI in the young. The effect of smoking on coronary thrombus formation may partially be attributed to its FXIII increasing effect.


Asunto(s)
Factor XIII , Polimorfismo Genético , Infarto del Miocardio con Elevación del ST , Fumar , Adulto , Factor XIII/genética , Factor XIII/metabolismo , Femenino , Humanos , Masculino , Factores de Riesgo , Infarto del Miocardio con Elevación del ST/genética , Infarto del Miocardio con Elevación del ST/metabolismo , Fumar/efectos adversos , Fumar/genética , Fumar/metabolismo
7.
J Transl Med ; 14: 74, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26980433

RESUMEN

BACKGROUND: Aspirin resistance established by different laboratory methods is still a debated problem. Using COX1 specific methods no aspirin resistance was detected among healthy volunteers. Here we tested the effect of chronic aspirin treatment on platelets from patients with stable coronary artery disease. The expression of COX2 mRNA in platelets and its influences on the effect of aspirin was also investigated. METHODS: One hundred and forty four patients were enrolled in the study. The direct measurement of COX1 acetylation was carried out by monoclonal antibodies specific to acetylated and non-acetylated COX1 (acCOX1 and nacCOX1) using Western blotting technique. Arachidonic acid (AA) induced TXB2 production by platelets was measured by competitive immunoassay. AA induced platelet aggregation, ATP secretion and VerifyNow Aspirin Assay were also performed. COX2 and COX1 mRNA expression in platelets were measured in 56 patients by RT-qPCR. RESULTS: In 138 patients only acCOX1 was detected, in the remaining six patients nacCOX1 disappeared after a compliance period. AA induced TXB2 production by platelets was very low in all patients including the 6 patients after compliance. AA induced platelet aggregation, secretion and with a few exceptions the VerifyNow Assay also demonstrated the effect of aspirin. Smoking, diabetes mellitus and inflammatory conditions did not influence the results. The very low amount of COX2 mRNA detected in 39 % of the investigated platelets did not influence the effect of aspirin. CONCLUSIONS: No aspirin resistance was detected among patients with stable coronary artery disease. COX2 expression in platelets did not influence the effect of aspirin.


Asunto(s)
Aspirina/farmacología , Enfermedad de la Arteria Coronaria , Resistencia a Medicamentos , Acetilación/efectos de los fármacos , Adulto , Anciano , Anciano de 80 o más Años , Ácido Araquidónico/farmacología , Aspirina/uso terapéutico , Enfermedad de la Arteria Coronaria/tratamiento farmacológico , Enfermedad de la Arteria Coronaria/prevención & control , Ciclooxigenasa 1/genética , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Agregación Plaquetaria/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Tromboxano B2/biosíntesis
8.
Semin Thromb Hemost ; 42(4): 429-39, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27071048

RESUMEN

Inherited deficiency of FXIII A subunit (FXIII-A) is a rare (1:2,000,000) but very severe bleeding diathesis. The incidence is much higher in communities where the practice of consanguineous marriage is combined with founder effect mutation. Because of the high risk of intracranial bleeding, life-long prophylaxis, preferably using FXIII concentrate, is mandatory. In FXIII-B subunit deficiency the bleeding diathesis is mild to moderate. FXIII deficiency is frequently associated with impaired wound healing. Women suffering from FXIII deficiency cannot carry pregnancies to term; in severe cases spontaneous abortion occurs in the first trimester. Plasma-derived heat-inactivated FXIII concentrate and recombinant FXIII-A are available for prophylaxis; a 4 weekly dose of 35 to 40 U/kg is recommended and a trough level of greater than 5% FXIII activity should be aimed for. During pregnancy, 2 weekly prophylaxis with a target trough level of greater than 10% is recommended, and during labor FXIII activity should exceed 30%. During surgical procedures, the target should be higher than 50% FXIII activity. Alloantibodies make FXIII deficiency difficult to manage, but fortunately they are extremely rare. Acquired FXIII deficiency may involve both subunits. Autoantibodies against FXIII subunits also manifest in severe bleeding complication with a relatively high mortality rate. The first-line test in the diagnosis of FXIII deficiency should be a quantitative functional assay based on the measurement of ammonia release or amine incorporation. The sensitivity of the traditional clot solubility assay is not sufficiently robust to enable proper screening. Antigen assays are needed for the classification of FXIII deficiencies. In the case of anti-FXIII antibodies, the diagnostic armory should be supplemented by a mixing test/Bethesda-type inhibitor assay and by assays that detect/measure the binding of antibodies to FXIII and to its subunits.


Asunto(s)
Enfermedades Autoinmunes , Deficiencia del Factor XIII , Hemorragia , Complicaciones Hematológicas del Embarazo , Autoanticuerpos/sangre , Enfermedades Autoinmunes/sangre , Enfermedades Autoinmunes/diagnóstico , Enfermedades Autoinmunes/terapia , Deficiencia del Factor XIII/sangre , Deficiencia del Factor XIII/complicaciones , Deficiencia del Factor XIII/diagnóstico , Deficiencia del Factor XIII/terapia , Femenino , Hemorragia/sangre , Hemorragia/diagnóstico , Hemorragia/etiología , Hemorragia/terapia , Humanos , Masculino , Embarazo , Complicaciones Hematológicas del Embarazo/sangre , Complicaciones Hematológicas del Embarazo/diagnóstico , Complicaciones Hematológicas del Embarazo/terapia
10.
Blood ; 123(11): 1757-63, 2014 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-24408323

RESUMEN

Coagulation factor XIII (FXIII) is a heterotetramer consisting of 2 catalytic A subunits (FXIII-A2) and 2 protective/inhibitory B subunits (FXIII-B2). FXIII-B, a mosaic protein consisting of 10 sushi domains, significantly prolongs the lifespan of catalytic subunits in the circulation and prevents their slow progressive activation in plasmatic conditions. In this study, the biochemistry of the interaction between the 2 FXIII subunits was investigated. Using a surface plasmon resonance technique and an enzyme-linked immunosorbent assay-type binding assay, the equilibrium dissociation constant (Kd) for the interaction was established in the range of 10(-10) M. Based on the measured Kd, it was calculated that in plasma approximately 1% of FXIII-A2 should be in free form. This value was confirmed experimentally by measuring FXIII-A2 in plasma samples immunodepleted of FXIII-A2B2. Free plasma FXIII-A2 is functionally active, and when activated by thrombin and Ca(2+), it can cross-link fibrin. In cerebrospinal fluid and tears with much lower FXIII subunit concentrations, >80% of FXIII-A2 existed in free form. A monoclonal anti-FXIII-B antibody that prevented the interaction between the 2 subunits reacted with the recombinant combined first and second sushi domains of FXIII-B, and its epitope was localized to the peptide spanning positions 96 to 103 in the second sushi domain.


Asunto(s)
Líquidos Corporales/química , Factor XIII/metabolismo , Factor XIIIa/metabolismo , Proteínas Recombinantes/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Formación de Anticuerpos , Reactivos de Enlaces Cruzados/farmacología , Ensayo de Inmunoadsorción Enzimática , Mapeo Epitopo , Factor XIII/inmunología , Factor XIIIa/inmunología , Fibrina/metabolismo , Humanos , Cinética , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Resonancia por Plasmón de Superficie
11.
Platelets ; 27(1): 59-65, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26083485

RESUMEN

Dual antiplatelet therapy with clopidogrel and aspirin is frequently used for the prevention of recurrent ischemic events. Various laboratory methods are used to detect the effect of these drugs administered in monotherapy, however their value in dual therapy has not been explored. Here, we determined which methods used for testing the effect of clopidogrel or aspirin are influenced by the other antiplatelet agent. One arm of the study included 53 ischemic stroke patients being on clopidogrel monotherapy showing effective inhibition of the P2Y12 ADP receptor. Laboratory tests routinely used for the detection of aspirin resistance (arachidonic acid (AA)-induced platelet aggregation/secretion, AA-induced thromboxane B2 (TXB2) production in platelet-rich plasma and VerifyNow Aspirin assay) were carried out on samples obtained from these patients. The other arm of the study involved 52 patients with coronary artery disease being on aspirin monotherapy. Methods used for testing the effect of clopidogrel (ADP-induced platelet aggregation and secretion, flow cytometric analysis of vasodilator-stimulated phosphoprotein (VASP) phosphorylation and a newly developed P2Y12-specific platelet aggregation (ADP[PGE1] test)) were performed on samples obtained from these patients. Clopidogrel monotherapy significantly inhibited AA-induced platelet aggregation and secretion, moreover, AA-induced TXB2 production was also significantly decreased. VASP phosphorylation and AA-induced platelet aggregation showed fair correlation in patients taking clopidogrel only. Clopidogrel did not inhibit the VerifyNow Aspirin test significantly. Aspirin monotherapy influenced ADP-induced platelet aggregation and secretion, but did not have an effect on VASP phosphorylation and on the ADP[PGE1] platelet aggregation test.


Asunto(s)
Aspirina/administración & dosificación , Inhibidores de Agregación Plaquetaria/administración & dosificación , Ticlopidina/análogos & derivados , Aspirina/sangre , Estudios de Casos y Controles , Clopidogrel , Interacciones Farmacológicas , Monitoreo de Drogas/métodos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/sangre , Ticlopidina/administración & dosificación , Ticlopidina/sangre
12.
Int J Mol Sci ; 16(1): 1143-59, 2015 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-25569091

RESUMEN

The aim of the case-control study was to explore the effect of coagulation factor XIII (FXIII) B subunit (FXIII-B) polymorphisms on the risk of coronary artery disease, and on FXIII levels. In the study, 687 patients admitted for coronary angiography to investigate suspected coronary artery disease and 994 individuals representing the Hungarian population were enrolled. The patients were classified according to the presence of significant coronary atherosclerosis (CAS) and history of myocardial infarction (MI). The F13B gene was genotyped for p.His95Arg and for intron K nt29756 C>G polymorphisms; the latter results in the replacement of 10 C-terminal amino acids by 25 novel amino acids. The p.His95Arg polymorphism did not influence the risk of CAS or MI. The FXIII-B intron K nt29756 G allele provided significant protection against CAS and MI in patients with a fibrinogen level in the upper tertile. However, this effect prevailed only in the presence of the FXIII-A Leu34 allele, and a synergism between the two polymorphisms was revealed. Carriers of the intron K nt29756 G allele had significantly lower FXIII levels, and FXIII levels in the lower tertile provided significant protection against MI. It is suggested that the protective effect of the combined polymorphisms is related to decreased FXIII levels.


Asunto(s)
Enfermedad de la Arteria Coronaria/genética , Factor XIII/genética , Polimorfismo de Nucleótido Simple , Anciano , Alelos , Estudios de Casos y Controles , Enfermedad de la Arteria Coronaria/complicaciones , Enfermedad de la Arteria Coronaria/patología , Factor XIII/análisis , Factor XIIIa/genética , Femenino , Fibrinógeno/análisis , Genotipo , Heterocigoto , Humanos , Intrones , Masculino , Persona de Mediana Edad , Infarto del Miocardio/complicaciones , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Oportunidad Relativa , Factores de Riesgo
13.
Clin Chem Lab Med ; 52(12): 1797-806, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24968404

RESUMEN

BACKGROUND: Antithrombin (AT) is a slow-acting progressive inhibitor of activated clotting factors, particularly thrombin and activated factor X (FXa). However, the presence of heparin or heparan sulfate accelerates its effect by several magnitudes. AT deficiency, a severe thrombophilia, is classified as type I (quantitative) and type II (qualitative) deficiency. In the latter case mutations may influence the reactive site, the heparin binding-site (HBS) and exert pleiotropic effect. Heterozygous type II-HBS deficiency is a less severe thrombophilia than other heterozygous subtypes. However, as opposed to other subtypes, it also exists in homozygous form which represents a very high risk of venous thromboembolism. METHODS: A modified anti-FXa chromogenic AT assay was developed which determines both the progressive (p) and the heparin cofactor (hc) activities, in parallel. The method was evaluated and reference intervals were established. The usefulness of the assay in detecting type II-HBS AT deficiency was tested on 78 AT deficient patients including 51 type II-HBS heterozygotes and 18 homozygotes. RESULTS: Both p-anti-FXa and hc-anti-FXa assays showed excellent reproducibility and were not influenced by high concentrations of triglyceride, bilirubin and hemoglobin. Reference intervals for p-anti-FXa and hc-anti-FXa AT activities were 84%-117% and 81%-117%, respectively. Type II-HBS deficient patients demonstrated low (heterozygotes) or very low (homozygotes) hc-anti-FXa activity with normal or slightly decreased p-anti-FXa activity. The p/hc ratio clearly distinguished wild type controls, type II-HBS heterozygotes and homozygotes. CONCLUSIONS: Concomitant determination of p-anti-FXa and hc-anti-FXa activities provides a reliable, clinically important diagnosis of type II-HBS AT deficiency and distinguishes between homozygotes and heterozygotes.


Asunto(s)
Deficiencia de Antitrombina III/diagnóstico , Antitrombina III/análisis , Factor Xa/metabolismo , Antitrombina III/genética , Antitrombina III/normas , Deficiencia de Antitrombina III/clasificación , Bilirrubina/química , Pruebas de Coagulación Sanguínea/normas , Factor Xa/química , Hemoglobinas/química , Heparina/química , Heterocigoto , Homocigoto , Humanos , Masculino , Polimorfismo de Nucleótido Simple , Valores de Referencia , Triglicéridos/química
14.
Int J Mol Sci ; 14(2): 3456-66, 2013 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-23389042

RESUMEN

RT-qPCR is the standard method for studying changes in relative transcript level in different experimental and clinical conditions and in different tissues. No validated reference genes have been reported for the normalization of transcript level in platelets. The very low level of platelet RNA and the elimination of leukocyte contamination represented special methodological difficulties. Our aims were to apply a simple technique to separate platelets for transcript level studies, and select the most stable reference genes for platelets from healthy individuals and from patients with the history of myocardial infarction. We developed a simple, straightforward method of platelet separation for RNA isolation. Platelet activation was inhibited by using acid-citrate-dextrose for anticoagulation and by prostaglandin E1. Leukocyte contamination was eliminated by three consecutive centrifugations. Samples prepared by this method were free of leukocytes, showed no inhibition in PCR reaction and no RNA degradation. The assay demands low blood volume, which complies with the requirements of everyday laboratory routine. Seventeen potential reference genes were investigated, but eight of them were excluded during optimization. The stability of the remaining genes, EEF2, EAR, ACTB, GAPDH, ANAPC5, OAZ1, HDGF, GNAS, and CFL1, were determined by four different descriptive statistics. GAPDH, GNAS, and ACTB were shown to be the most stable genes in platelets of healthy individuals, while HDGF, GNAS, and ACTB were the most stable in platelets of patients with the history of myocardial infarction. The results confirm that data normalization needs assessment of appropriate reference genes for a particular sample set.

15.
Clin Chem Lab Med ; 50(7): 1191-202, 2012 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-22850052

RESUMEN

Coagulation factor XIII (FXIII) is converted by thrombin and Ca(2+) into an active transglutaminase (FXIIIa) in the final phase of coagulation cascade. Its main function is the mechanical stabilization of fibrin clot and its protection from fibrinolysis by cross-linking of fibrin chains and α(2)-plasmin inhibitor to fibrin. In non-substituted patients FXIII deficiency is a severe hemorrhagic diathesis, not infrequently with fatal consequences. The main reason for using FXIII assays is the diagnosis of FXIII deficiency. The aim of this review is to provide a comprehensive critical evaluation of the methods reported for the determination of FXIII activity in the plasma. Such methods are based on two principles: 1) measurement of labeled amines incorporated by FXIIIa into a glutamine residue of a substrate protein, 2) monitoring ammonia released from a peptide bound glutamine residue by FXIIIa using NAD(P)H dependent glutamate dehydrogenase indicator reaction. The incorporation assays are sensitive, but cumbersome and time-consuming, they are difficult to standardize and cannot be automated. The ammonia release assays are less sensitive, but quick, well standardized, and can be automated; this type of assay is recommended for the screening of FXIII deficiency. The traditional clot solubility assay should not be used for this purpose.


Asunto(s)
Deficiencia del Factor XIII/sangre , Factor XIII/metabolismo , Factor XIII/análisis , Humanos , Trombina/metabolismo , Trombosis/sangre
16.
Curr Opin Hematol ; 18(5): 366-72, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21738029

RESUMEN

PURPOSE OF REVIEW: Here we review recent developments concerning the diagnosis, classification and treatment of factor XIII (FXIII) deficiency and new findings related to the pathogenesis of the disease. RECENT FINDINGS: Most recently, the International Society on Thrombosis and Haemostasis, Scientific and Standardization Committee published a guideline for the diagnosis and classification of FXIII deficiencies. Since 2009, three novel mutations causing severe bleeding diathesis were discovered in the FXIII-A gene and one in the FXIII-B gene. A newly described FXIII-A deficiency was of the extremely rare qualitative type II deficiency. The first well established founder effect was reported for a causative FXIII-A mutation. More than a quarter of all FXIII-A deficiencies are due to autoantibody, among them the first case of deficiency caused by anti-FXIII-B autoantibody was reported in the last 2 years. The safety and effectiveness of plasma FXIII concentrate for prophylaxis and treatment is now well established. The new recombinant FXIII product is currently in phase III clinical trial and the preliminary data are promising. SUMMARY: FXIII deficiency is considered the most underdiagnosed bleeding diathesis. The recommended algorithm for its diagnosis and classification could improve the diagnostic efficiency. The preferred choice for substitution therapy is FXIII concentrate (plasma-derived or, in the future, recombinant).


Asunto(s)
Deficiencia del Factor XIII , Deficiencia del Factor XIII/clasificación , Deficiencia del Factor XIII/diagnóstico , Deficiencia del Factor XIII/terapia , Humanos
17.
J Thromb Haemost ; 20(2): 525-531, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34784091

RESUMEN

BACKGROUND: Factor XIII (FXIII)-B subunit measurements are required for the diagnosis and characterization of the type of FXIII deficiency. Furthermore, therapy for FXIII-A deficiency with recombinant FXIII (rFXIII-A) relies on available FXIII-B. OBJECTIVE: To carry out a collaborative study to calibrate and assign value to the current WHO 1st International Standard (IS) FXIII Plasma for Total FXIII-B subunit, relative to locally collected normal plasma pools. METHODS: Laboratories were instructed to use a validated method (specific ELISA antibodies provided) for assessment of Total FXIII-B subunit antigen potency. All laboratories used this method with one laboratory using an additional in-house method. Nine data sets were received from seven laboratories (37 assays in total), which provided a total of 35 valid estimates for this new assignment. Total FXIII-B subunit estimates were calculated relative to locally collected normal plasma pools, using an arbitrary value of 1.00 unit of Total FXIII-B subunit per ml, for each pool. RESULTS: Combination of results produced an overall mean of 0.98 units/mL with an inter-laboratory variability (geometric coefficients of variation - GCV%) of 18.3% [95% confidence interval: 0.86-1.11]. Real-time and bench stability studies indicated good stability and preservation of the FXIII-B subunit analyte in the WHO 1st IS FXIII Plasma (02/206). CONCLUSION: Following agreement by study participants, ISTH/SSC Experts, WHO-ISTH Liaison Group and the SSC Board, the WHO/ECBS established the current WHO 1st IS Factor XIII plasma (NIBSC code 02/206) by additionally assigning it with a Total FXIII-B subunit antigen value of 0.98 IU/ampoule, in October 2019.


Asunto(s)
Deficiencia del Factor XIII , Factor XIII , Factor XIII/análisis , Deficiencia del Factor XIII/diagnóstico , Fibrinógeno , Humanos , Organización Mundial de la Salud
18.
J Thromb Haemost ; 20(5): 1223-1235, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35146910

RESUMEN

BACKGROUND: Platelets contain a high amount of potentially active A subunit dimer of coagulation factor XIII (cellular FXIII; cFXIII). It is of cytoplasmic localization, not secreted, but becomes translocated to the surface of platelets activated by convulxin and thrombin (CVX+Thr). OBJECTIVE: To explore the difference in cFXIII translocation between receptor mediated and non-receptor mediated platelet activation and if translocation can also be detected on platelet-derived microparticles. Our aim was also to shed some light on the mechanism of cFXIII translocation. METHODS: Gel-filtered platelets were activated by CVX+Thr or Ca2+ -ionophore (calcimycin). The translocation of cFXIII and phosphatidylserine (PS) to the surface of activated platelets and platelet-derived microparticles was investigated by flow cytometry, immunofluorescence, and immune electron microscopy. Fluo-4-AM fluorescence was used for the measurement of intracellular Ca2+ concentration. RESULTS: Receptor mediated activation by CVX+Thr exposed cFXIII to the surface of more than 60% of platelets. Electron microscopy revealed microparticles with preserved membrane structure and microparticles devoid of labeling for membrane glycoprotein CD41a. cFXIII was observed on both types of microparticles but was more abundant in the absence of CD41a. Rhosin, a RhoA inhibitor, significantly decreased cFXIII translocation. Non-receptor mediated activation of platelets by calcimycin elevated intracellular Ca2+ concentration, induced the translocation of PS to the surface of platelets and microparticles, but failed to expose cFXIII. CONCLUSIONS: The elevation of intracellular Ca2+ concentration is sufficient for the translocation of PS from the internal layer of the membrane, while the translocation of cFXIII from the platelet cytoplasm requires additional receptor mediated mechanism(s).


Asunto(s)
Micropartículas Derivadas de Células , Factor XIII , Plaquetas , Calcimicina/farmacología , Proteínas Portadoras , Humanos , Fosfatidilserinas , Activación Plaquetaria , Trombina/farmacología
19.
Semin Thromb Hemost ; 37(3): 305-14, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21455864

RESUMEN

Plasma factor XIII (FXIII) is a tetrameric zymogen consisting of two potentially active A subunits (FXIII-A) and two carrier/inhibitory B subunits (FXIII-B). In the final phase of the coagulation cascade, FXIII is converted into an active transglutaminase (FXIIIa) by thrombin and Ca (2 + ). FXIIIa strengthens fibrin clot mechanically by cross-linking fibrin chains. In addition, FXIIIa is a key regulator of fibrinolysis, protecting newly formed fibrin from the fibrinolytic machinery by binding α (2)-plasmin inhibitor to the fibrin meshwork. FXIII is essential for maintaining hemostasis; its severe deficiency causes a life-threatening bleeding diathesis. The involvement of FXIII in thrombotic diseases and its association with the risk of these disorders is less clear. The role of FXIII in atherothrombotic diseases has been recently reviewed. This article offers a general overview of the relationship between FXIII and venous thromboembolism (VTE), to collect individual publications on this topic, present conclusions, and examine limitations of published studies. Special attention is given to the association of FXIII-A polymorphism with the risk of VTE, which has provoked considerable interest over the last decade.


Asunto(s)
Factor XIII/genética , Tromboembolia Venosa/genética , Factor XIII/metabolismo , Factor XIIIa/genética , Fibrina/metabolismo , Humanos , Polimorfismo Genético
20.
Blood ; 113(3): 723-5, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-18955560

RESUMEN

Acquired factor XIII (FXIII) deficiency due to autoantibody against FXIII is a very rare severe hemorrhagic diathesis. Antibodies directed against the A subunit of FXIII, which interfere with different functions of FXIII, have been described. Here, for the first time, we report an autoantibody against the B subunit of FXIII (FXIII-B) that caused life-threatening bleeding in a patient with systemic lupus erythematosus. FXIII activity, FXIII-A(2)B(2) complex, and individual FXIII subunits were undetectable in the plasma, whereas platelet FXIII activity and antigen were normal. Neither FXIII activation nor its activity was inhibited by the antibody, which bound to structural epitope(s) on both free and complexed FXIII-B. The autoantibody highly accelerated the elimination of FXIII from the circulation. FXIII supplementation combined with immunosuppressive therapy, plasmapheresis, immunoglobulin, and anti-CD20 treatment resulted in the patient's recovery. FXIII levels returned to around 20% at discharge and after gradual increase the levels stabilized above 50%.


Asunto(s)
Autoanticuerpos/inmunología , Autoantígenos/inmunología , Deficiencia del Factor XIII/inmunología , Factor XIII/inmunología , Hemorragia/inmunología , Adulto , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales de Origen Murino , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Factor XIII/análisis , Deficiencia del Factor XIII/terapia , Femenino , Humanos , Inmunoglobulinas Intravenosas , Factores Inmunológicos/uso terapéutico , Inmunosupresores/uso terapéutico , Fallo Renal Crónico/etiología , Lupus Eritematoso Sistémico/complicaciones , Plasmaféresis , Diálisis Renal , Rituximab
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA