Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Blood ; 123(26): 4045-53, 2014 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-24829206

RESUMEN

Ex vivo gene therapy strategies avoid systemic delivery of viruses thereby mitigating the risk of vector-associated immunogenicity. Previously, we delivered autologous factor VIII (FVIII)-expressing blood outgrowth endothelial cells (BOECs) to hemophilia A mice and showed that these cells remained sequestered within the implanted matrix and provided therapeutic levels of FVIII. Prior to translating this strategy into the canine (c) model of hemophilia A, we increased cFVIII transgene expression by at least 100-fold with the use of the elongation factor 1 alpha (EF1α) promoter and a strong endothelial enhancer element. BOECs isolated from hemophilia A dogs transduced with this lentiviral vector express levels of cFVIII ranging between 1.0 and 1.5 U/mL per 10(6) cells over 24 hours. Autologous BOECs have been implanted into the omentum of 2 normal and 3 hemophilia A dogs. These implanted cells formed new vessels in the omentum. All 3 hemophilia A dogs treated with FVIII-expressing autologous BOECs developed anti-FVIII immunoglobulin G2 antibodies, but in only 2 of the dogs were these antibodies inhibitory. FVIII antigen levels >40% in the absence of FVIII coagulant function were detected in the circulation for up to a year after a single gene therapy treatment, indicating prolonged cellular viability and synthesis of FVIII.


Asunto(s)
Autoanticuerpos , Inhibidores de Factor de Coagulación Sanguínea , Células Endoteliales , Factor VIII , Expresión Génica , Terapia Genética/métodos , Hemofilia A , Animales , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Autoinjertos , Inhibidores de Factor de Coagulación Sanguínea/sangre , Inhibidores de Factor de Coagulación Sanguínea/inmunología , Modelos Animales de Enfermedad , Perros , Células Endoteliales/metabolismo , Células Endoteliales/trasplante , Factor VIII/biosíntesis , Factor VIII/genética , Factor VIII/inmunología , Vectores Genéticos , Hemofilia A/sangre , Hemofilia A/genética , Hemofilia A/inmunología , Hemofilia A/terapia , Humanos , Lentivirus , Ratones , Factor 1 de Elongación Peptídica/genética , Regiones Promotoras Genéticas , Transducción Genética , Transgenes
2.
Blood ; 121(26): 5228-37, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23529928

RESUMEN

Genetic variation in or near the C-type lectin domain family 4 member M (CLEC4M) has been associated with plasma levels of von Willebrand factor (VWF) in healthy individuals. CLEC4M is a lectin receptor with a polymorphic extracellular neck region possessing a variable number of tandem repeats (VNTR). A total of 491 participants (318 patients with type 1 von Willebrand disease [VWD] and 173 unaffected family members) were genotyped for the CLEC4M VNTR polymorphism. Family-based association analysis on kindreds with type 1 VWD demonstrated an excess transmission of VNTR 6 to unaffected individuals (P = .0096) and an association of this allele with increased VWF:RCo (P = .029). CLEC4M-Fc bound to VWF. Immunofluorescence and enzyme-linked immunosorbent assay demonstrated that HEK 293 cells transfected with CLEC4M bound and internalized VWF. Cells expressing 4 or 9 copies of the CLEC4M neck region VNTR showed reduced interaction with VWF relative to CLEC4M with 7 VNTR (CLEC4M 4%-60% reduction, P < .001; CLEC4M 9%-45% reduction, P = .006). Mice expressing CLEC4M after hydrodynamic liver transfer have a 46% decrease in plasma levels of VWF (P = .0094). CLEC4M binds to and internalizes VWF, and polymorphisms in the CLEC4M gene contribute to variable plasma levels of VWF.


Asunto(s)
Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Repeticiones de Minisatélite/genética , Polimorfismo Genético/genética , Enfermedades de von Willebrand/sangre , Factor de von Willebrand/metabolismo , Adolescente , Adulto , Animales , Estudios de Casos y Controles , Niño , Preescolar , ADN/genética , Ensayo de Inmunoadsorción Enzimática , Familia , Femenino , Citometría de Flujo , Estudio de Asociación del Genoma Completo , Genotipo , Células HEK293 , Humanos , Técnicas para Inmunoenzimas , Lactante , Desequilibrio de Ligamiento , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Adulto Joven , Enfermedades de von Willebrand/genética , Enfermedades de von Willebrand/patología
3.
J Thromb Haemost ; 22(1): 76-89, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37678547

RESUMEN

BACKGROUND: One-third of patients with severe hemophilia A develop neutralizing antibodies to the factor VIII (FVIII) protein in response to intravenous replacement therapy. Patients may also generate natural, nonneutralizing antibodies to FVIII before FVIII exposure. These patients are at increased risk of developing neutralizing antibodies to FVIII. However, natural anti-FVIII antibodies are also present in healthy human donors. OBJECTIVES: To further characterize the natural antihuman (h) FVIII antibody repertoire in mice and humans. METHODS: An in-house ELISA was developed using a purified polyclonal immunoglobulin (Ig) standard to quantify anti-hFVIII Ig in cell culture supernatant or plasma from mice (wild-type and FVIII-/-) and adult human donors. RESULTS: All naïve wild-type and FVIII-/- mice, as well as healthy human donors, possess natural anti-hFVIII antibodies. Mice only have natural anti-hFVIII IgM, which is present in germ-free mice, suggesting that they are germline encoded. Although murine marginal zone B cells (MZBs) contribute 44% to all circulating natural IgM, they contribute disproportionately to the anti-hFVIII IgM repertoire (82%). This naturally occurring murine MZB-derived IgM is not B-domain specific and is reduced by intravenously administered hFVIII, suggesting that it may form immune complexes immediately upon hFVIII administration. Natural anti-hFVIII antibodies of IgG, IgM, and IgA isotypes can be detected in adult human donors. There were increased levels of B-domain-favoring anti-hFVIII IgG in 14% of healthy donors, which were markedly different from the rest of the "low-titer" population. CONCLUSIONS: There is a preponderance of natural anti-hFVIII antibodies in both mice and healthy adult human donors.


Asunto(s)
Factor VIII , Hemofilia A , Adulto , Humanos , Ratones , Animales , Factor VIII/metabolismo , Inmunoglobulina G , Anticuerpos Neutralizantes , Inmunoglobulina M
4.
J Thromb Haemost ; 22(3): 666-675, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38040335

RESUMEN

BACKGROUND: As knowledge of the human genome has advanced, so too has the recognition that interpretation of the pathogenic nature of sequence variants can be challenging. The von Willebrand factor (VWF) gene exhibits a significant degree of sequence variability, and the first VWF variant associated with type 1 von Willebrand disease (VWD), c.4751 A>G, p.Y1584C, was described in 2003. However, since that time, the pathogenic nature of this variant has remained unclear, being assigned properties ranging from a risk factor to a pathogenic variant. OBJECTIVES: To provide additional evaluation on the interpretation of pathogenicity for this common VWF variant. METHODS: Fifty-eight subjects with only the p.Y1584C variant were recruited from 2 cohort studies (the Zimmerman Program and the Canadian type 1 VWD study). Clinical and laboratory phenotypes were assessed. RESULTS: The prevalence of the p.Y1584C variant in our cohorts was 23- to 27-fold higher than that in large normal population databases. Significantly more p.Y1584C subjects had an abnormal bleeding score when compared to Y1584 individuals. In comparison with a group of 35 subjects without the p.Y1584C variant, subjects with the variant had lower mean VWF:antigen and VWF:ristocetin cofactor values and significantly higher VWF propeptide/VWF:antigen ratios suggestive of enhanced clearance. CONCLUSION: Collectively, the results of this analysis suggest that p.Y1584C is likely pathogenic, however, due to influences such as incomplete penetrance, variable expressivity, and other genetic modifiers like ABO blood group, the straightforward assignment of pathogenicity to this variant is inevitably challenging.


Asunto(s)
Enfermedad de von Willebrand Tipo 1 , Enfermedades de von Willebrand , Humanos , Factor de von Willebrand/genética , Factor de von Willebrand/análisis , Canadá , Enfermedad de von Willebrand Tipo 1/diagnóstico , Fenotipo , Enfermedades de von Willebrand/diagnóstico , Enfermedades de von Willebrand/genética
5.
Blood ; 117(16): 4358-66, 2011 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-21346256

RESUMEN

Type 1 VWD is the mild to moderate reduction of VWF levels. This study examined the mechanisms underlying 2 common type 1 VWD mutations, the severe R1205H and more moderate Y1584C. In vitro biosynthesis was reduced for both mutations in human and mouse VWF, with the effect being more severe in R1205H. VWF knockout mice received hydrodynamic injections of mouse Vwf cDNA. Lower VWF antigen levels were demonstrated in both homozygous and heterozygous forms for both type 1 mutations from days 14-42. Recombinant protein infusions and hydrodynamic-expressed VWF propeptide to antigen ratios demonstrate that R1205H mouse VWF has an increased clearance rate, while Y1584C is normal. Recombinant ADAMTS13 digestions of Y1584C demonstrated enhanced cleavage of both human and mouse VWF115 substrates. Hydrodynamic-expressed VWF shows a loss of high molecular weight multimers for Y1584C compared with wild-type and R1205H. At normal physiologic levels of VWF, Y1584C showed reduced thrombus formation in a ferric chloride injury model while R1205H demonstrated similar thrombogenic activity to wild-type VWF. This study has elucidated several novel mechanisms for these mutations and highlights that the type 1 VWD phenotype can be recapitulated in the VWF knockout hydrodynamic injection model.


Asunto(s)
Mutación , Enfermedades de von Willebrand/genética , Factor de von Willebrand/genética , Proteínas ADAM/metabolismo , Proteína ADAMTS13 , Animales , Línea Celular , Modelos Animales de Enfermedad , Humanos , Metaloendopeptidasas/metabolismo , Ratones , Ratones Noqueados , Fenotipo , Multimerización de Proteína , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Trombosis/genética , Factor de von Willebrand/administración & dosificación , Factor de von Willebrand/metabolismo
6.
Blood ; 116(18): 3645-52, 2010 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-20696945

RESUMEN

We have studied the effect of a 13-bp deletion in the promoter of the von Willebrand factor (VWF) gene in a patient with type 1 von Willebrand disease. The index case has a VWF:Ag of 0.49 IU/mL and is heterozygous for the deletion. The deletion is located 48 bp 5' of the transcription start site, and in silico analysis, electrophoretic mobility shift assays, and chromatin immunoprecipitation studies all predict aberrant binding of Ets transcription factors to the site of the deletion. Transduction of reporter gene constructs into blood outgrowth endothelial cells showed a 50.5% reduction in expression with the mutant promoter (n = 16, P < .001). A similar 40% loss of transactivation was documented in transduced HepG2 cells. A similar marked reduction of transgene expression was shown in the livers of mice injected with the mutant promoter construct (n = 8, P = .003). Finally, in studies of BOEC mRNA, the index case showed a 4.6-fold reduction of expression of the VWF transcript associated with the deletion mutation. These studies show that the 13-bp deletion mutation alters the binding of Ets (and possibly GATA) proteins to the VWF promoter and significantly reduces VWF expression, thus playing a central pathogenic role in the type 1 von Willebrand disease phenotype in the index case.


Asunto(s)
Regiones Promotoras Genéticas , Eliminación de Secuencia , Activación Transcripcional , Enfermedad de von Willebrand Tipo 1/genética , Factor de von Willebrand/genética , Animales , Línea Celular , Células Endoteliales/metabolismo , Factores de Transcripción GATA/metabolismo , Células Hep G2 , Humanos , Ratones , Ratones Endogámicos BALB C , Fenotipo , Unión Proteica , Proteínas Proto-Oncogénicas c-ets/metabolismo , ARN Mensajero/genética , Factores de Transcripción/metabolismo , Transgenes , Enfermedad de von Willebrand Tipo 1/metabolismo , Factor de von Willebrand/metabolismo
7.
J Thromb Haemost ; 20(11): 2507-2518, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35950488

RESUMEN

BACKGROUND: Substantial phenotypic heterogeneity exists in endothelial cells and while much of this heterogeneity results from local microenvironments, epigenetic modifications also contribute. METHODS: Cultured human umbilical vein endothelial cells, human pulmonary microvascular endothelial cells, human hepatic sinusoidal endothelial cells, human lymphatic endothelial cells (hLECs), and two different isolations of endothelial colony forming cells (ECFCs) were assessed for levels of factor VIII (FVIII) and von Willebrand factor (VWF) RNA and protein. The intracellular location and co-localization of both proteins was evaluated with immunofluorescence microscopy and stimulated release toof FVIII and VWF from Weibel-Palade bodies (WPBs) was evaluated. Changes in expression of FVIII and VWF RNA after hLECs and ECFCs were exposed to 2 or 15 dynes/cm2 of laminar shear stress were also assessed. RESULTS: We observed considerable heterogeneity in FVIII and VWF expression among the endothelial cells. With the exception of hLECs, FVIII RNA and protein were barely detectable in any of the endothelial cells and a reciprocal relationship between levels of FVIII and VWF appears to exist. When FVIII and VWF are co-expressed, they do not consistently co-localize in the cytoplasm. However, in hLECs where significantly higher levels of FVIII are expressed, FVIII and VWF co-localize in WPBs and are released together when stimulated. Expression of both FVIII and VWF is markedly reduced when hLECs are exposed to higher or lower levels of laminar shear stress, while in ECFCs there is a minimal response for both proteins. CONCLUSIONS: Variable levels of FVIII and VWF RNA and protein exist in a subset of cultured human endothelial cells. Higher levels of FVIII present in hLECs co-localize with VWF and are released together when exposed to a secretagogue.


Asunto(s)
Hemostáticos , Factor de von Willebrand , Humanos , Factor de von Willebrand/metabolismo , Factor VIII/genética , Factor VIII/metabolismo , Secretagogos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , ARN
8.
J Thromb Haemost ; 19(10): 2440-2453, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34152080

RESUMEN

BACKGROUND: Stabilin-2 is an endocytic scavenger receptor that mediates the clearance of glycosaminoglycans, phosphatidylserine-expressing cells, and the von Willebrand factor-factor VIII (FVIII) complex. In a genome-wide screening study, pathogenic loss-of-function variants in the human STAB2 gene associated with an increased incidence of unprovoked venous thromboembolism (VTE). However, the specific mechanism(s) by which stabilin-2 deficiency influences the pathogenesis of VTE is unknown. OBJECTIVES: The aim of this study was to assess the influence of stabilin-2 on deep vein thrombosis (DVT) and to characterize the underlying prothrombotic phenotype of stabilin-2 deficiency in a mouse model. METHODS: DVT was induced using the inferior vena cava (IVC) stenosis model in two independent cohorts (littermates and non-littermates) of wild-type (Stab2+/+ ) and stabilin-2 (Stab2-/- )-deficient mice. Thrombus structure and contents were quantified by immunohistochemistry. Plasma procoagulant activity was assessed and complete blood counts were performed. RESULTS: Incidence of thrombus formation was not altered between Stab2+/+ and Stab2-/- mice. When thrombi were formed, Stab2-/- mice developed significantly larger thrombi than Stab2+/+ controls. Thrombi from Stab2-/- mice contained significantly more leukocytes and citrullinated histone H3 than Stab2+/+ thrombi. Stab2-/- mice had increased FVIII activity. Circulating levels of monocytes and granulocytes were significantly elevated in Stab2-/- mice, and Stab2-/- mice had elevated plasma cell-free DNA 24 hours post-IVC stenosis compared to their Stab2+/+ counterparts. CONCLUSIONS: These data suggest that stabilin-2 deficiency associates with a prothrombotic phenotype involving elevated levels of neutrophil extracellular trap-releasing leukocytes coupled with endogenous procoagulant activity, resulting in larger and qualitatively distinct venous thrombi.


Asunto(s)
Trampas Extracelulares , Trombosis , Animales , Moléculas de Adhesión Celular Neuronal , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Venas , Factor de von Willebrand
9.
J Thromb Haemost ; 18(10): 2721-2731, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32654420

RESUMEN

BACKGROUND: Endothelial colony forming cells (ECFCs) derived from peripheral blood can be used to analyze the pathophysiology of vascular diseases ex vivo. However, heterogeneity is observed between ECFC clones and this variability needs to be understood and standardized for ECFCs to be used as a cell model for applications in vascular studies. OBJECTIVE: Determine reference characteristics of healthy control ECFCs to generate a valid ex vivo model for vascular disease. METHODS: Putative ECFCs (n = 47) derived from 21 individual healthy subjects were studied for cell morphology and specific cell characteristics. Clones were analyzed for the production and secretion of von Willebrand factor (VWF), cell proliferation, and the expression of endothelial cell markers. RESULTS: Based on morphology, clones were categorized into three groups. Group 1 consisted of clones with classic endothelial cell morphology, whereas groups 2 and 3 contained less condensed cells with increasing cell sizes. All clones had comparable endothelial cell surface expression profiles, with low levels of non-endothelial markers. However, a decrease in CD31 and a group-related increase in CD309 and CD45 expression, combined with a decrease in cell proliferation and VWF production and secretion, was observed in clones in group 3 and to a lesser extent in group 2. CONCLUSIONS: We observed group-related variations in endothelial cell characteristics when clones lacked the classic endothelial cell morphology. Despite this variation, clones in all groups expressed endothelial cell surface markers. Provided that clones with similar characteristics are compared, we believe ECFCs are a valid ex vivo model to study vascular disease.


Asunto(s)
Células Endoteliales , Enfermedades Vasculares , Membrana Celular , Proliferación Celular , Células Cultivadas , Estado de Salud , Humanos , Neovascularización Fisiológica , Factor de von Willebrand
10.
J Thromb Haemost ; 17(4): 681-694, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30740857

RESUMEN

Essentials CLEC4M is an endocytic receptor for factor FVIII. CLEC4M interacts with FVIII in a VWF-dependent and independent manner. CLEC4M binds to mannose-containing glycans on FVIII. CLEC4M internalization of FVIII involves clathrin coated pits. SUMMARY: Background von Willebrand factor (VWF) and factor VIII (FVIII) circulate in the plasma as a non-covalent complex, and the majority of FVIII is likely to be cleared by VWF-dependent pathways. Clearance of VWF-free FVIII is rapid and underlies the pathological basis of some quantitative FVIII deficiencies. The receptor pathways that regulate the clearance of VWF-bound and VWF-free FVIII are incompletely uncharacterized. The human liver-expressed endothelial lectin CLEC4M has been previously characterized as a clearance receptor for VWF, although its influence on FVIII is unknown. Objective The interaction between FVIII and CLEC4M was characterized in the presence or absence of VWF. Methods FVIII interactions with CLEC4M were evaluated by in vitro cell-based and solid phase binding assays. Interactions between FVIII and CLEC4M or liver sinusoidal endothelial cells were evaluated in vivo by immunohistochemistry. Results CLEC4M-expressing HEK 293 cells bound and internalized recombinant and plasma-derived FVIII through VWF-dependent and independent mechanisms. CLEC4M binding to recombinant FVIII was dependent on mannose-exposed N-linked glycans. CLEC4M mediated FVIII internalization via a clathrin-coated pit-dependent mechanism, resulting in transport of FVIII from early and late endosomes for catabolism by lysosomes. In vivo hepatic expression of CLEC4M after hydrodynamic liver transfer was associated with a decrease in plasma levels of endogenous murine FVIII:C in normal mice, whereas infused recombinant human FVIII was associated with sinusoidal endothelial cells in the presence or absence of VWF. Conclusions These findings suggest that CLEC4M is a novel clearance receptor that interacts with mannose-exposed glycans on FVIII in the presence or absence of VWF.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Endocitosis , Células Endoteliales/metabolismo , Factor VIII/metabolismo , Lectinas Tipo C/metabolismo , Hígado/irrigación sanguínea , Receptores de Superficie Celular/metabolismo , Factor de von Willebrand/metabolismo , Animales , Sitios de Unión , Moléculas de Adhesión Celular/genética , Clatrina/metabolismo , Endosomas/metabolismo , Factor VIII/genética , Células HEK293 , Humanos , Lectinas Tipo C/genética , Lisosomas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , Transporte de Proteínas , Proteolisis , Receptores de Superficie Celular/genética , Factor de von Willebrand/genética
11.
J Thromb Haemost ; 17(8): 1384-1396, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31126000

RESUMEN

BACKGROUND: Scavenger receptors play a significant role in clearing aged proteins from the plasma, including the large glycoprotein coagulation factors von Willebrand factor (VWF) and factor VIII (FVIII). A large genome-wide association study meta-analysis has identified genetic variants in the gene SCARA5, which encodes the class A scavenger receptor SCARA5, as being associated with plasma levels of VWF and FVIII. OBJECTIVES: The ability of SCARA5 to regulate the clearance of VWF-FVIII was characterized. METHODS: VWF-FVIII interactions with SCARA5 were evaluated by solid phase binding assays and in vitro cell based assays. The influence of SCARA5 deficiency on VWF:Ag and half-life was assessed in a murine model. The expression pattern of SCARA5 and its colocalization with VWF was evaluated in human tissues. RESULTS: VWF and the VWF-FVIII complex bound to human recombinant SCARA5 in a dose- and calcium-dependent manner. SCARA5 expressing HEK 293T cells bound and internalized VWF and the VWF-FVIII complex into early endosomes. In vivo, SCARA5 deficiency had a modest influence on the half-life of human VWF. mRNA analysis and immunohistochemistry determined that human SCARA5 is expressed in kidney podocytes and the red pulp, white pulp, and marginal zone of the spleen. VWF was found to colocalize with SCARA5 expressed by littoral cells lining the red pulp of the human spleen. CONCLUSIONS: SCARA5 is an adhesive and endocytic receptor for VWF. In human tissues, SCARA5 is expressed by kidney podocytes and splenic littoral endothelial cells. SCARA5 may have a modest influence on VWF clearance in humans.


Asunto(s)
Endocitosis , Receptores Depuradores de Clase A/metabolismo , Bazo/metabolismo , Factor de von Willebrand/metabolismo , Animales , Factor VIII/metabolismo , Femenino , Células HEK293 , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Podocitos/metabolismo , Unión Proteica , Receptores Depuradores de Clase A/genética , Bazo/citología
12.
Br J Haematol ; 143(4): 552-8, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18986390

RESUMEN

The multimeric plasma protein von Willebrand factor (VWF) is regulated in size by its protease, ADAMTS13 (a disintegrin and metalloproteinase with thrombospondin type 1 motif, member 13). Y1605-M1606 cleavage site mutations and single nucleotide polymorphisms (SNPs) in the VWF A1 and A2 domains were examined for alteration in ADAMTS13-mediated cleavage of VWF. Recombinant human full-length VWF (rVWF) was digested with recombinant human ADAMTS13 (rADAMTS13) using a dialysis membrane method with 1.5 mol/l urea, and analyzed via multimer migration distance. The glutathione-S-transferase (GST) and histidine-tagged construct, E1554-R1668 of VWF (VWF115) was assayed via enzyme-linked immunosorbent assay: VWF115 was bound to anti-GST coated plates, digested with rADAMTS13, and intact VWF115 detected via horseradish peroxidase-labelled anti-histidine tag antibody. All alterations examined in the Y1605-M1606 cleavage site greatly reduced the cleavability of VWF by ADAMTS13 in the rVWF assay. Greatest cleavage resistance in both assays was observed in Y1605A/M1606A. In contrast, Y1605H and M1606L show a loss of cleavability only in the rVWF assay, suggesting that an aromatic ring at 1605 is critical for ADAMTS13 recognition. Additionally, under our rVWF assay conditions, the G1643S polymorphism showed increased cleavage, suggesting a Type 2A VWD phenotype, while D1472H, Q1571H and P1601T showed slightly decreased ADAMTS13 cleavage. Our two complementary assay conditions show that A-domain changes in VWF alter ADAMTS13-mediated proteolysis.


Asunto(s)
Proteínas ADAM/genética , Factor de von Willebrand/genética , Proteínas ADAM/sangre , Proteína ADAMTS13 , División del ADN , Humanos , Mutagénesis , Polimorfismo de Nucleótido Simple , Estructura Terciaria de Proteína/genética , Proteínas Recombinantes/metabolismo , Factor de von Willebrand/metabolismo
13.
J Clin Invest ; 128(9): 4057-4073, 2018 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-30124466

RESUMEN

Quantitative abnormalities of the von Willebrand factor-factor VIII (VWF-FVIII) complex associate with inherited bleeding or thrombotic disorders. Receptor-mediated interactions between plasma VWF-FVIII and phagocytic or immune cells can influence their hemostatic and immunogenic activities. Genetic association studies have demonstrated that variants in the STAB2 gene, which encodes the scavenger receptor stabilin-2, associate with plasma levels of VWF-FVIII. However, the mechanistic basis and pathophysiological consequences of this association are unknown. We have demonstrated that stabilin-2-expressing cells bind and internalize human VWF and FVIII in a VWF-dependent manner, and stabilin-2-deficient mice displayed prolonged human VWF-FVIII half-life compared with controls. The stabilin-2 variant p.E2377K significantly decreased stabilin-2 expression and impaired VWF endocytosis in a heterologous expression system, and common STAB2 variants associated with plasma VWF levels in type 1 von Willebrand disease patients. STAB2-deficient mice displayed a decreased immunogenic response to human VWF-FVIII complex, while coinfusion of human VWF-FVIII with the stabilin-2 ligand hyaluronic acid attenuated the immune response to exogenous FVIII. Collectively, these data suggest that stabilin-2 functions as both a clearance and an immunoregulatory receptor for VWF-FVIII, making stabilin-2 a novel molecular target for modification of the half-life of VWF-FVIII and the immune response to VWF-FVIII concentrates.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Factor VIII/metabolismo , Factor de von Willebrand/metabolismo , Adolescente , Adulto , Anciano , Animales , Moléculas de Adhesión Celular Neuronal/deficiencia , Niño , Preescolar , Combinación de Medicamentos , Endocitosis , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Factor VIII/química , Factor VIII/inmunología , Factor VIII/farmacocinética , Femenino , Variación Genética , Semivida , Humanos , Lactante , Hígado/citología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Unión Proteica , Estabilidad Proteica , Adulto Joven , Factor de von Willebrand/química , Factor de von Willebrand/inmunología , Factor de von Willebrand/farmacocinética
14.
Hum Gene Ther ; 13(13): 1583-93, 2002 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12228013

RESUMEN

If gene therapy is to be an effective treatment modality for hemophilia A, therapeutic levels and tissue-restricted expression of factor VIII (FVIII) must be achieved through optimization of transgene expression. To this end, we incorporated three types of sequence elements into a canine B domain-deleted FVIII transgene cassette and individually evaluated their effect on FVIII transgene expression. Functional FVIII activity was initially assessed in vitro and hydrodynamic injection of the different transgene constructs into mice was subsequently used as a model to compare in vivo expression of the various modified transgenes. Our results demonstrate that in vitro transgene expression is, in these studies, not a good predictor of in vivo transgene performance. In vivo analysis of a hybrid tissue-restricted promoter element, consisting of a concatemer of five hepatocyte nuclear factor 1 (HNF-1) consensus-binding motifs juxtaposed to the human FVIII proximal promoter, indicates that it is as efficient at mediating expression of the FVIII protein as the cytomegalovirus promoter. Addition of the full-length canine FVIII 3'-UTR also enhances transgene expression of FVIII in vivo. Sequence analysis of the canine FVIII 3'-UTR and human FVIII 3'-UTR indicates that the former lacks instability sequences and may therefore be more effective in stabilizing FVIII mRNA. Subsequent inclusion of FVIII introns 16 and 17 into the natural locations of the transgene disrupted mRNA processing and abolished expression of the FVIII protein. Introduction of intron 17 proximal to the FVIII cDNA did not enhance in vivo expression of canine FVIII from the transgene.


Asunto(s)
Regiones no Traducidas 3'/genética , Factor VIII/genética , Terapia Genética , Hemofilia A/terapia , Factores de Transcripción/metabolismo , Animales , Perros , Factor VIII/metabolismo , Hemofilia A/genética , Intrones , Factores de Transcripción/genética , Transgenes
15.
Thromb Haemost ; 87(4): 659-65, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12008949

RESUMEN

We have identified the causative mutation in the hemophilia A dog colony at Queen's University, Canada and have observed a striking similarity with the intron 22 inversion found in approximately 45% of severely affected hemophilia A patients. The canine hemophilia A phenotype arises from aberrant splicing and premature termination of transcription of the FVIII gene, resulting in a polyadenylated transcript lacking exons distal to 22 and terminating with a novel sequence element (NSE). In dogs and other species including humans, this NSE is present in low copy number. One copy of these sequences in the canine genome is within intron 22 and reveals differences in the hybridization banding patterns between normal and hemophilic DNA, suggestive of a large genomic rearrangement. The mutation mechanism may not be uncommon, as identical mutant transcripts were isolated from two hemophilia A littermates that are unrelated to the Queen's colony and from hemophiliac dogs in the colony at Chapel Hill.


Asunto(s)
Inversión Cromosómica , Enfermedades de los Perros/genética , Perros/genética , Factor VIII/genética , Hemofilia A/veterinaria , Empalme del ARN , Animales , Secuencia de Consenso , Análisis Mutacional de ADN , ADN Complementario/genética , Exones/genética , Biblioteca de Genes , Hemofilia A/genética , Humanos , Intrones/genética , Masculino , Poliadenilación , Polimorfismo Genético , Sitios de Empalme de ARN/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especificidad de la Especie
16.
Thromb Haemost ; 105(3): 501-8, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21301777

RESUMEN

Less than 50 patients are reported with platelet type von Willebrand disease (PT-VWD) worldwide. Several reports have discussed the diagnostic challenge of this disease versus the closely similar disorder type 2B VWD. However, no systematic study has evaluated this dilemma globally. Over three years, a total of 110 samples/data from eight countries were analysed. A molecular approach was utilised, analysing exon 28 of the von Willebrand factor (VWF) gene, and in mutation negative cases the platelet GP1BA gene. Our results show that 48 cases initially diagnosed as putative type 2B/PT-VWD carried exon 28 mutations consistent with type 2B VWD, 17 carried GP1BA mutations consistent with a PT-VWD diagnosis, three had other VWD types (2A and 2M) and five expressed three non-previously published exon 28 mutations. Excluding 10 unaffected family members and one acquired VWD, 26 cases did not have mutations in either genes. Based on our study, the percentage of type 2B VWD diagnosis is 44% while the percentage of misdiagnosis of PT-VWD is 15%. This is the first large international study to investigate the occurrence of PT-VWD and type 2B VWD worldwide and to evaluate DNA analysis as a diagnostic tool for a large cohort of patients. The study highlights the diagnostic limitations due to unavailability/poor application of RIPA and related tests in some centres and proposes genetic analysis as a suitable tool for the discrimination of the two disorders worldwide. Cases that are negative for both VWF and GP1BA gene mutations require further evaluation for alternative diagnoses.


Asunto(s)
Plaquetas/metabolismo , Enfermedad de von Willebrand Tipo 2/sangre , Factor de von Willebrand/biosíntesis , Trastornos de las Plaquetas Sanguíneas/genética , Plaquetas/citología , ADN/metabolismo , Exones , Femenino , Hemostasis , Humanos , Cooperación Internacional , Masculino , Glicoproteínas de Membrana/genética , Mutación , Recuento de Plaquetas , Complejo GPIb-IX de Glicoproteína Plaquetaria , Sistema de Registros , Enfermedad de von Willebrand Tipo 2/epidemiología
17.
Blood ; 109(1): 145-54, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17190853

RESUMEN

In order to evaluate the changes within the VWF gene that might contribute to the pathogenesis of type 1 von Willebrand disease (VWD), a large multicenter Canadian study was undertaken. We present data from the sequence analysis of the VWF gene in 123 type 1 VWD index cases and their families. We have identified putative mutations within the VWF gene in 63% (n = 78) of index cases, leaving 37% (n = 45) with no identified changes. These changes comprise 50 different putative mutations: 31 (62%) missense mutations, 8 (16%) changes involving the VWF transcriptional regulatory region, 5 (10%) small deletions/insertions, 5 (10%) splicing consensus sequence mutations, and 1 nonsense mutation. Twenty-one of the index cases had more than one putative VWF mutation identified. We were somewhat more likely to identify putative mutations in cases with lower VWF levels, and the contribution of other factors, such as ABO blood group, seems more important in milder cases. Taken as a whole, our data support a complex spectrum of molecular pathology resulting in type 1 VWD. In more severe cases, genetic changes are common within the VWF gene and are highly penetrant. In milder cases, the genetic determinants are more complex and involve factors outside of the VWF gene.


Asunto(s)
Mutación , Enfermedades de von Willebrand/genética , Factor de von Willebrand/genética , Sistema del Grupo Sanguíneo ABO/genética , Adolescente , Adulto , Anciano , Sustitución de Aminoácidos , Canadá/epidemiología , Niño , Preescolar , Estudios de Cohortes , Análisis Mutacional de ADN , Salud de la Familia , Femenino , Frecuencia de los Genes , Genotipo , Humanos , Lactante , Masculino , Persona de Mediana Edad , Mutación Missense , Fenotipo , Mutación Puntual , Enfermedades de von Willebrand/sangre , Enfermedades de von Willebrand/clasificación , Enfermedades de von Willebrand/epidemiología , Factor de von Willebrand/análisis
18.
Blood ; 105(4): 1531-9, 2005 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-15498853

RESUMEN

Mechanisms of tissue-restricted patterns of von Willebrand factor (VWF) expression involve activators and repressors that limit expression to endothelial cells and megakaryocytes. The relative transcriptional activity of the proximal VWF promoter was assessed in VWF-producing and -nonproducing cells, and promoter activity was highest in endothelial cells followed by megakaryocytes. Only basal VWF promoter activity was seen in nonendothelial cells. Here we identify a negative response element located at nucleotides (nts) +96/+105 and demonstrate, using chromatin immunoprecipitation (ChIP) analysis, that in vivo this sequence interacts with the E4BP4 transcriptional repressor. Differences in size and relative abundance of nuclear E4BP4 were observed. In HepG2 cells, low levels of larger forms of E4BP4 are present that directly interact with the negative response element. In VWF-expressing cells, high levels of smaller forms predominate with no evidence of direct DNA binding. However, in endothelial cells, mutation of the VWF E4BP4 binding motif not only restores but also further elevates VWF promoter activity, suggesting that E4BP4 may be part of a coordinated binding complex. These observations implicate this binding motif in repressing both activated and basal levels of VWF transcription by different cell type-specific mechanisms, and support the hypothesis that E4BP4 sequesters negative regulators of transcription, thereby enhancing activated gene expression.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Proteínas Represoras/fisiología , Factores de Transcripción/fisiología , Factor de von Willebrand/biosíntesis , Factor de von Willebrand/metabolismo , Animales , Secuencia de Bases , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico , Western Blotting , Bovinos , Línea Celular , Línea Celular Tumoral , Cricetinae , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Exones/genética , Factores de Unión a la G-Box , Células HeLa , Humanos , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Proteínas Nucleares/análisis , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transfección , Factor de von Willebrand/antagonistas & inhibidores , Factor de von Willebrand/genética
19.
Blood ; 105(11): 4330-6, 2005 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15705799

RESUMEN

Interaction between the platelet glycoprotein Ibalpha (GPIbalpha) receptor and its adhesive ligand von Willebrand factor (VWF) has a critical role in the process of hemostasis. Platelet-type von Willebrand disease (PT-VWD) is a rare bleeding disorder that results from gain-of-function mutations in the GPIBA gene. We studied this gene from 5 members of a previously unreported family with a PT-VWD phenotype. We identified a novel in-frame deletion of 27 base pair (bp) in the macroglycopeptide region. This deletion was not found in the unaffected family members or in 50 healthy controls. The patients' platelets expressed normal quantities of GPIb/IX/V complex on their surface and the mutant (Mut) GPIbalpha was expressed at levels indistinguishable from the wild-type (WT) receptor on the surface of transfected Chinese hamster ovary (CHO) beta/IX cells. Analysis of ristocetin-mediated (125)I-VWF binding showed that the Mut receptor binds VWF in the absence of ristocetin and displays an increased sensitivity to lower concentrations of the modulator. This is the first report of a gain-of-function mutation in the GPIbalpha receptor outside the VWF-binding domain in patients with PT-VWD. The mutation provides a molecular basis for the PT-VWD phenotype and supports a role for the macroglycopeptide region in receptor function.


Asunto(s)
Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Eliminación de Secuencia , Enfermedades de von Willebrand/genética , Adulto , Secuencia de Bases , Plaquetas , Células Cultivadas , Salud de la Familia , Femenino , Humanos , Linaje , Fenotipo , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Unión Proteica , Transfección , Enfermedades de von Willebrand/etiología , Factor de von Willebrand/metabolismo
20.
Blood ; 104(9): 2739-45, 2004 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-15226181

RESUMEN

In this manuscript, we describe a case of type 2A von Willebrand disease (VWD) caused by the novel heterozygous G>A transition at nucleotide 3538, which should result in the putative, nonconservative substitution of G1180R. This mutation was reproduced by site-directed mutagenesis; however, the recombinant mutant protein was efficiently secreted from cells and assembled correctly into multimers. Because the substitution is located at the last nucleotide of exon 26, the patient's platelet von Willebrand factor (VWF) mRNA was analyzed and 3 transcripts were observed: the normal transcript without the 3538G>A transition, a transcript with the in-frame deletion of exon 26, and a transcript with the in-frame deletions of exons 23 and 26. These deletion VWF cDNA constructs were created and the resulting recombinant proteins were analyzed following transfection into COS-7 cells. Cotransfection results demonstrate that the exon-skipped transcripts led to intracellular retention, and the levels of VWF antigen (VWF:Ag) produced by these constructs were as follows: del23/26A transition at nucleotide (nt) 3538 does not result in the expression of the G1180R missense mutation, but rather leads to exon skipping, which is the pathogenic basis of the patient's phenotype. This is the first report of a coding region mutation resulting in the skipping of 2 nonadjacent exons.


Asunto(s)
Exones , Mutación Puntual/fisiología , Eliminación de Secuencia , Factor de von Willebrand/genética , Adolescente , Secuencia de Aminoácidos , Salud de la Familia , Femenino , Humanos , ARN/genética , Enfermedades de von Willebrand/etiología , Enfermedades de von Willebrand/genética , Factor de von Willebrand/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA