Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Front Immunol ; 11: 563, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32457734

RESUMEN

Objectives: In hemophilia A the presence of non-neutralizing antibodies (NNAs) against Factor VIII (FVIII) may predict the development of neutralizing antibodies (inhibitors) and accelerate the clearance of administrated FVIII concentrates. This systematic review aimed to assess: (1) the prevalence and incidence of NNAs in patients with congenital hemophilia without inhibitors and (2) the association between NNAs and patient and treatment characteristics. Methods: We conducted a search in MEDLINE, Embase, Web of Science and the Cochrane database. We included cross-sectional and longitudinal studies reporting on NNAs in patients with hemophilia A and B, who were inhibitor-negative at the start of the observation period. Data were extracted on: hemophilia type and severity, patient and treatment characteristics, NNA prevalence and incidence, NNA assays and inhibitor development. Two independent reviewers performed study selection, data extraction and risk of bias assessment, using adapted criteria of the Joanna Briggs Institute. Studies were classified as high-quality when ≥5/9 criteria were met. NNA assays were classified as high-quality when both quality criteria were met: (1) use of positive controls and (2) competition with FVIII to establish FVIII-specificity. We reported NNA prevalence and incidence for each study. The pooled NNA prevalence was assessed for well-designed studies in previously treated patients, employing high-quality NNA assays. Results: We included data from 2,723 inhibitor-negative patients with hemophilia A, derived from 28 studies. Most studies were cross-sectional (19/28) and none reported on NNAs in hemophilia B. Study design was of high quality in 16/28 studies and the NNA assay quality was high in 9/28 studies. Various NNA assays were used, predominantly ELISA (18/28) with different cut-off values. We found a large variety in NNA prevalence (Range, 0-100%). The pooled NNA prevalence in high-quality studies was 25% (95% CI, 16-38%). The incidence of new NNA development was reported in one study (0.01 NNA per person-exposure day). Conclusion: This systematic review identified studies that were heterogeneous in study design, patient population and NNA assay type, with NNA prevalence ranging from 0 to 100% in inhibitor-negative patients with hemophilia A. The pooled NNA prevalence was 25% in high-quality studies including only previously treated patients and performing high-quality NNA assays.


Asunto(s)
Autoanticuerpos/inmunología , Factor VIII/inmunología , Hemofilia A/inmunología , Factor VIII/uso terapéutico , Femenino , Hemofilia A/tratamiento farmacológico , Humanos , Incidencia , Masculino , Prevalencia
2.
Mol Genet Metab ; 126(2): 162-168, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30473480

RESUMEN

BACKGROUND: Treatment of Fabry disease (FD) with recombinant alpha-galactosidase A (r-αGAL A) is complicated by the formation of anti-drug antibodies in the majority of male patients with the classical disease phenotype. Detailed information regarding antibody subtypes, onset and persistence of antibody development and their effect on treatment efficacy is sparse. METHODS: A retrospective study was carried out in 39 male patients with classical FD, treated with either agalsidase-alfa or agalsidase-beta (mean follow up of 10 years). With six to twelve months intervals plasma-induced in vitro inhibition of enzyme activity, lysoglobotriaosylsphingosine (lysoGb3) levels and renal function were assessed. In a subset of 12 patients, additionally anti- r-αGAL A IgM, IgA and IgG1, 2, 3 and 4 levels were analyzed. RESULTS: In 23 out of 39 patients, plasma-induced in vitro inhibition of r-αGAL A activity was observed (inhibition-positive). The inhibition titer was strongly negatively correlated to the decrease in lysoGb3: agalsidase-alfa (FElog10(inhibition) = -10.3, P ≤.001), agalsidase-beta (FElog10(inhibition) = -4.7, P ≤.001). Inhibition-positive patients had an accelerated decline in renal function (FE = 1.21, p = .042). During treatment IgG1 anti-r-αGAL A levels increased only in inhibition-positive patients (p = .0045). IgG4 anti-r-αGAL A antibodies developed in 7 out of 9 inhibition-positive patients. Other antibody subclasses were either not present or too low to quantify. CONCLUSION: Development of inhibiting antibodies against r-αGAL A negatively affects the biochemical response to ERT and resulted in an accelerated decline in renal function. The presence of IgG1 and IgG4 anti-r-αGAL A antibodies is associated with in vitro αGAL A activity inhibition.


Asunto(s)
Anticuerpos/clasificación , Enfermedad de Fabry/tratamiento farmacológico , Isoenzimas/inmunología , Proteínas Recombinantes/inmunología , alfa-Galactosidasa/inmunología , Adolescente , Adulto , Anticuerpos/inmunología , Estudios de Seguimiento , Humanos , Inmunoglobulina G/inmunología , Isoenzimas/uso terapéutico , Masculino , Persona de Mediana Edad , Proteínas Recombinantes/uso terapéutico , Estudios Retrospectivos , Resultado del Tratamiento , Adulto Joven , alfa-Galactosidasa/uso terapéutico
3.
J Thromb Haemost ; 16(3): 533-545, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29285874

RESUMEN

Essentials Initial immune cell interactions leading to factor (F) VIII immunity are not well characterized. We assessed cellular interactions and expression profiles in hemophilia A mice. MARCO+, followed by SIGLEC1+ and SIGNR1+ macrophages co-localize most with human FVIII. The splenic transcriptome highlights potential therapeutic targets to prevent inhibitors. SUMMARY: Background Developing factor VIII (FVIII) inhibitory antibodies is the most serious complication in hemophilia A treatment, representing a significant health and economic burden. A better understanding of the early events in an immune response leading to this outcome may provide insight into inhibitor development. Objective To identify early mediators of FVIII immunity and to detail immune expression profiles in the spleen and liver. Methods C57Bl/6 F8 E16 knockout mice were infused with 5-20 µg (2000-8000 IU kg-1 ) of recombinant FVIII. Spleens were frozen at various time-points post-infusion and stained for FVIII and cellular markers. Splenic and liver RNA expression analysis was performed 3 h post-infusion of 0.6 µg (240 IU kg-1 ) FVIII by nCounter technology using a 561-gene immunology panel. Results FVIII localization in the spleen did not change over 2.5 h. We observed significantly higher co-localization of FVIII with MARCO+ cells compared with SIGLEC1+ and SIGNR1+ in the splenic marginal zone. FVIII exhibited little co-localization with CD11c+ dendritic cells and the macrophage mannose receptor, CD206. Following FVIII infusion, the splenic mRNA profiling identified genes such as Tnfaip6 and Il23r, which are implicated in chemotaxis and a proinflammatory Th17 response, respectively. In contrast, an upregulation of Gfi1 in the liver suggests an anti-inflammatory environment. Conclusions FVIII co-localizes predominantly with marginal zone macrophages (MARCO+ ) in the murine spleen following intravenous infusion. Targeting pathways that are implicated in the early FVIII innate immune response in the spleen may lead to therapeutic interventions to prevent inhibitor formation.


Asunto(s)
Factor VIII/metabolismo , Regulación de la Expresión Génica , Hemofilia A/genética , Hemofilia A/inmunología , Transcriptoma , Animales , Moléculas de Adhesión Celular/metabolismo , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Sistema Inmunológico , Inmunohistoquímica , Lectinas Tipo C/metabolismo , Hígado/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , Receptores de Superficie Celular/metabolismo , Lectina 1 Similar a Ig de Unión al Ácido Siálico/metabolismo , Bazo/metabolismo
4.
J Thromb Haemost ; 16(2): 378-388, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29222940

RESUMEN

Essentials Conformational changes in ADAMTS-13 are part of its mode-of-action. The murine anti-ADAMTS-13 antibody 1C4 discriminates between folded and open ADAMTS-13. ADAMTS-13 conformation is open in acute acquired thrombotic thrombocytopenic purpura (TTP). Our study forms an important basis to fully elucidate the pathophysiology of TTP. SUMMARY: Background Acquired thrombotic thrombocytopenic purpura (aTTP) is an autoimmune disorder characterized by absent ADAMTS-13 activity and the presence of anti-ADAMTS-13 autoantibodies. Recently, it was shown that ADAMTS-13 adopts a folded or an open conformation. Objectives As conformational changes in self-antigens play a role in the pathophysiology of different autoimmune diseases, we hypothesized that the conformation of ADAMTS-13 changes during acute aTTP. Methods Antibodies recognizing cryptic epitopes in the spacer domain were generated. Next, the conformation of ADAMTS-13 in 40 healthy donors (HDs), 99 aTTP patients (63 in the acute phase versus 36 in remission), 12 hemolytic-uremic syndrome (HUS) patients and 63 sepsis patients was determined with ELISA. Results The antibody 1C4 recognizes a cryptic epitope in ADAMTS-13. Therefore, we were able to discriminate between a folded and an open ADAMTS-13 conformation. We showed that ADAMTS-13 in HDs does not bind to 1C4, indicating that ADAMTS-13 circulates in a folded conformation. Similar results were obtained for HUS and sepsis patients. In contrast, ADAMTS-13 of acute aTTP patients bound to 1C4 in 92% of the cases, whereas, in most cases, this binding was abolished during remission, showing that the conformation of ADAMTS-13 is open during an acute aTTP episode. Conclusions Our study shows that, besides absent ADAMTS-13 activity and the presence of anti-ADAMTS-13 autoantibodies, an open ADAMTS-13 conformation is also a hallmark of acute aTTP. Demonstrating this altered ADAMTS-13 conformation in acute aTTP will help to further unravel the pathophysiology of aTTP and lead to improved therapy and diagnosis.


Asunto(s)
Proteína ADAMTS13/química , Púrpura Trombocitopénica Trombótica/enzimología , Proteína ADAMTS13/sangre , Proteína ADAMTS13/inmunología , Autoanticuerpos/inmunología , Autoanticuerpos/metabolismo , Estudios de Casos y Controles , Ensayo de Inmunoadsorción Enzimática , Mapeo Epitopo , Epítopos , Humanos , Unión Proteica , Conformación Proteica , Pliegue de Proteína , Púrpura Trombocitopénica Trombótica/sangre , Púrpura Trombocitopénica Trombótica/diagnóstico , Púrpura Trombocitopénica Trombótica/inmunología , Relación Estructura-Actividad
5.
J Thromb Haemost ; 15(2): 246-254, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27943580

RESUMEN

Essentials Factor VIII levels vary in mild and moderate hemophilia A (MHA) patients with the same mutation. We aimed to estimate the variation and determinants of factor VIII levels among MHA patients. Age and genotype explain 59% of the observed inter-individual variation in factor VIII levels. Intra-individual variation accounted for 45% of the variation in the three largest mutation groups. SUMMARY: Background The bleeding phenotype in patients with mild/moderate hemophilia A (MHA) is inversely associated with the residual plasma concentration of factor VIII (FVIII:C). Within a group of patients with the same F8 missense mutation, baseline FVIII:C may vary, because, in healthy individuals, von Willebrand factor (VWF) levels, ABO blood group and age are also known to influence baseline FVIII:C. Our understanding of the pathophysiologic process of the causative genetic event leading to reduced baseline FVIII:C in MHA patients is still limited. Objectives To estimate the variation and determinants of baseline FVIII:C among MHA patients with the same F8 missense mutation. Methods Three hundred and forty-six patients carrying mutations that were present in at least 10 patients in the cohort were selected from the INSIGHT and the RISE studies, which are cohort studies including data of 3534 MHA patients from Europe, Canada, and Australia. Baseline FVIII:C was measured with a one-stage clotting assay. We used Levene's test, univariate and multivariate linear regression, and mixed-model analyses. Results For 59% of patients, the observed variation in baseline FVIII:C was explained by age and genotype. Compared to FVIII:C in patients with Arg612Cys, FVIII:C was significantly different in patients with eight other F8 missense mutations. Intra-individual variation explained 45% of the observed variance in baseline FVIII:C among patients with the same mutation. Conclusion Our results indicate that baseline FVIII:C levels are not exclusively determined by F8 genotype in MHA patients. Insights into other factors may provide potential novel targets for the treatment of MHA.


Asunto(s)
Factor VIII/análisis , Hemofilia A/genética , Hemofilia A/metabolismo , Mutación , Sistema del Grupo Sanguíneo ABO , Adulto , Coagulación Sanguínea , Desamino Arginina Vasopresina/química , Factor VIII/genética , Variación Genética , Genotipo , Hemorragia , Humanos , Masculino , Persona de Mediana Edad , Análisis Multivariante , Mutación Missense , Variaciones Dependientes del Observador , Fenotipo , Conformación Proteica , Estudios Retrospectivos , Adulto Joven , Factor de von Willebrand/metabolismo
6.
J Thromb Haemost ; 15(2): 329-340, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27868337

RESUMEN

Essentials Anti-factor (F) VIII antibody formation is a major complication in the treatment of hemophilia A. We investigated uptake of FVIII and FVIII immune complex by bone marrow derived dendritic cells. Immune complex formation increased uptake of FVIII 3-4 fold in a Fcγ receptor dependent manner. FVIII immune complex binding to Fcγ receptors may modulate immune tolerance induction. SUMMARY: Background A major complication in the treatment of hemophilia A is the development of inhibitory antibodies targeting coagulation factor VIII (FVIII). Eradication of these inhibitors can be established by immune tolerance induction (ITI), which consists of daily administration of high dosages of FVIII. FVIII immune complexes (FVIII-IC) could be formed following FVIII infusion in patients with pre-existing anti-FVIII antibodies. Objectives Here we studied endocytosis of FVIII-IC by bone marrow-derived dendritic cells (BMDCs). Methods BMDCs were pulsed with FVIII/FVIII-IC and uptake was assessed by flow cytometry and confocal imaging. Results BMDCs were able to efficiently internalize FVIII-IC in a dose-dependent manner, 3-4-fold more efficiently when compared with equimolar concentrations of non-complexed FVIII. Uptake of FVIII-IC, but not FVIII alone, could be inhibited with anti-Fcγ receptor (FcγR) antibody 2.4G2, indicating functional involvement of FcγR. No internalization of FVIII-IC was observed in BMDCs lacking FcγRI, FcγRIIb, FcγRIII and FcγRIV. Genetic ablation of FcγRIIb, FcγRIII or FcγRIV individually did not affect the ability of anti-FVIII IgG to promote the uptake of FVIII. BMDCs lacking FcγRI showed lower FVIII-IC uptake levels when compared with other single FcγR null BMDCs. Expression of the inhibitory FcγRIIb alone was sufficient to internalize FVIII-IC more efficiently than FVIII. Conclusions FcγR are critical in the internalization of FVIII-IC by BMDCs and multiple FcγR can contribute independently to this process. Our findings provide a basis for future studies to address whether the outcome of ITI is dependent on the interplay between FVIII-IC and inhibitory and activating FcγR.


Asunto(s)
Células Presentadoras de Antígenos/metabolismo , Factor VIII/metabolismo , Hemofilia A/terapia , Animales , Anticuerpos Monoclonales/administración & dosificación , Complejo Antígeno-Anticuerpo/inmunología , Células Presentadoras de Antígenos/inmunología , Coagulación Sanguínea , Células de la Médula Ósea/metabolismo , Células Dendríticas/metabolismo , Endocitosis , Factor VIII/inmunología , Hemofilia A/inmunología , Humanos , Tolerancia Inmunológica , Inmunoglobulina G/química , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Conformación Molecular , Ratas , Receptores de IgG/metabolismo , Proteínas Recombinantes/metabolismo
7.
Vox Sang ; 111(3): 247-256, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27184018

RESUMEN

BACKGROUND AND OBJECTIVES: In mice, loss of sialic acid resulting in shedding of glycoprotein (GP) Ibα and GPV has been linked to platelet survival. The aim of this study was to determine whether loss of sialic acid and the GPIb-IX-V complex contributes to development of the platelet storage lesion (PSL) in human platelet concentrates (PCs). MATERIALS AND METHODS: PCs (stored in plasma (with or without Mirasol treatment); PAS-C or PAS-E) were stored at room temperature. Flow cytometry was used to monitor membrane expression of the GPIb-IX-V complex, CD62P, surface glycans and PS exposure. The functionality of stored platelets was determined employing aggregometry and ristocetin-induced VWF binding. RESULTS: Storage time of PCs in blood banks is limited to 7 days. During this time period, a minor but gradually increasing subpopulation of GPIbα-negative platelets was observed. Also, ristocetin-induced VWF binding was impaired in a small population of platelets. Mean surface expression of GPIbα and GPV remained stable until day 9, whereas CD62P expression increased; also a rapid decrease in ADP-induced aggregation was observed for PAS-C, PAS-E and Mirasol-treated PCs. Upon prolonged storage (>9 days), a slow decline in surface expression of GPIbα and GPV was observed; no major changes were observed in surface sialylation with the exception of Mirasol-treated platelets. CONCLUSION: In a small population of stored platelets, changes in GPIbα occur from day 2 onwards. Loss of sialic acid and subsequent shedding of GPIbα and GPV is not an early event during the development of the PSL.


Asunto(s)
Plaquetas/metabolismo , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Plaquetas/citología , Plaquetas/efectos de los fármacos , Conservación de la Sangre , Crioprotectores/farmacología , Citometría de Flujo , Humanos , Ácido N-Acetilneuramínico/metabolismo , Selectina-P/metabolismo , Unión Proteica , Ristocetina/farmacología , Factor de von Willebrand/química , Factor de von Willebrand/metabolismo
8.
J Thromb Haemost ; 13(10): 1854-66, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26270243

RESUMEN

BACKGROUND: Endothelial von Willebrand factor (VWF) inhibits angiogenesis. Accordingly, blood outgrowth endothelial cells (BOECs) isolated from von Willebrand disease (VWD) patients showed enhanced in vitro angiogenesis when compared with healthy control BOECs. Characterization of the angiogenic response of VWD BOECs is limited and differences between the different types of VWD have not been investigated in detail. OBJECTIVES: The aim of this study was to further explore the potential pathogenic effect of VWF mutations on angiogenesis. METHODS: BOECs were isolated from four healthy individuals, 10 patients with VWD and one heterozygous carrier of a type 2N mutation. Cell migration and tube formation were measured. RESULTS: Migration velocity and total tube formation were similar between VWD patients and controls in general. BOECs from the type 3 VWD patient and one type 2B patient showed increased migratory velocity and tube formation compared with BOECs from other patients and healthy controls. Directional migration was impaired in eight out of 10 VWD BOECs and the ability to form tubes was limited to early passage numbers, but not for BOECs from healthy controls. CONCLUSION: BOECs can be a useful tool for ex vivo assessment of endothelial cell function in patients with different types of VWD, but possible limitations, such as early loss of angiogenic capacity, should be recognized. BOECs from most VWD patients consistently showed impairment in the directionality of migration. This is the first report on angiogenic properties of a type 3 VWD BOEC, which showed increased in vitro angiogenesis.


Asunto(s)
Células Endoteliales/metabolismo , Neovascularización Fisiológica , Enfermedades de von Willebrand/sangre , Factor de von Willebrand/metabolismo , Estudios de Casos y Controles , Movimiento Celular , Separación Celular , Células Cultivadas , Predisposición Genética a la Enfermedad , Heterocigoto , Humanos , Mutación , Neovascularización Fisiológica/genética , Fenotipo , Transducción de Señal , Enfermedades de von Willebrand/genética , Enfermedades de von Willebrand/fisiopatología , Factor de von Willebrand/genética
9.
J Thromb Haemost ; 12(5): 670-9, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24977290

RESUMEN

BACKGROUND: Acquired deficiency of ADAMTS13 causes a rare and life-threatening disorder called thrombotic thrombocytopenic purpura (TTP). Several studies have shown that aberrant glycosylation can play an important role in the pathogenesis of autoimmune diseases.N-linked glycosylation and putative O-fucosylation sites have been predicted or identified in recombinant ADAMTS13. However, it is not known which of these sites are glycosylated in plasma derived ADAMTS13. OBJECTIVES: Here we investigated the presence of putative O-fucosylation, C-mannosylation and N-linked glycosylation sites on plasma derived ADAMTS13. METHODS/RESULTS: Sites of N-linked glycosylation were determined by the use of peptide N-glycosidase-F (PNGase F), which removes the entire carbohydrate from the side chain of asparagines. Nine of the 10 predicted N-linked glycosylation sites were identified in or near the metalloproteinase,spacer, thrombospondin type 1 repeat (TSR1) and the CUB domain of plasma ADAMTS13. Moreover, six putative O-fucosylated sites were identified in the TSR domains of plasma ADAMTS13 by performing searches of the tandem mass spectrometry (MS/MS) data for loss of hexose (162 Da), deoxyhexose (146 Da), or hexose deoxyhexose(308 Da). The use of electron transfer dissociation (ETD) allowed for unambiguous identification of the modified sites. In addition to putative O-fucosylation and N-linked glycosylation, two putative C-mannosylation sites were identified within the TSR1 and TSR4 domains of ADAMTS13. CONCLUSIONS: Our data identify several glycosylation sites on plasma derived ADAMTS13. We anticipate that our findings may be relevant for the initiation of autoimmune reactivity against ADAMTS13 in patients with acquired TTP.


Asunto(s)
Proteínas ADAM/sangre , Púrpura Trombocitopénica Trombótica/sangre , Proteínas ADAM/genética , Proteína ADAMTS13 , Secuencia de Aminoácidos , Enfermedades Autoinmunes/inmunología , Fucosa/química , Glicosilación , Células HEK293 , Hexosas/química , Humanos , Manosa/química , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Púrpura Trombocitopénica Trombótica/genética , Homología de Secuencia de Aminoácido , Espectrometría de Masas en Tándem , Trombospondinas/sangre
10.
J Thromb Haemost ; 12(2): 273-81, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24283667

RESUMEN

BACKGROUND: Weibel-Palade bodies (WPBs) function as storage vesicles for von Willebrand factor (VWF) and a number of other bioactive compounds, including angiopoietin-2 and insulin-like growth factor-binding protein 7. WPBs release their content following stimulation with agonists that increase the level of intracellular Ca²âº, such as thrombin, or agonists that increase intracellular levels of cAMP, such as epinephrine. OBJECTIVE: Previously, we have shown that the exchange protein activated by cAMP, exchange protein activated by cAMP, and the small GTPase Rap1 are involved in cAMP-mediated release of WPBs. In this study, we explored potential downstream effectors of Rap1 in cAMP-mediated WPB release. METHODS: Studies were performed in primary human umbilical vein endothelial cells. Activation of the small GTP-binding protein Rac1 was monitored by its ability to bind to the CRIB domain of the serine/threonine kinase P21-activated kinase (PAK)1. Downstream effectors of Rap1 were identified with a proteomic screen using a glutathione-S-transferase fusion of the Ras-binding domain of RalGDS. Functional involvement of candidate proteins in WPB release was determined by RNA interference (RNAi)-mediated knockdown of gene expression. RESULTS: Depletion of Rac1 by RNAi prevented epinephrine-induced VWF secretion. Also, the Rac1 inhibitor EHT1864 reduced epinephrine-induced WPB release. We identified the phosphatidylinositol-3,4,5-triphosphate-dependent Rac exchange factor 1 (PREX1) and the regulatory ß-subunit of phosphatidylinositol 3-kinase (PI3K) as downstream targets of Rap1. The PI3K inhibitor LY294002 reduced epinephrine-induced release of VWF. RNAi-mediated downregulation of PREX1 abolished epinephrine-induced but not thrombin-induced release of WPBs. CONCLUSION: Our findings show that PREX1 regulates epinephrine-induced release of WPBs.


Asunto(s)
Epinefrina/farmacología , Exocitosis/efectos de los fármacos , Fosfatos de Fosfatidilinositol/metabolismo , Cuerpos de Weibel-Palade/efectos de los fármacos , Proteína de Unión al GTP rac1/metabolismo , AMP Cíclico/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Transducción de Señal , Cuerpos de Weibel-Palade/metabolismo , Factor de von Willebrand/metabolismo
11.
J Thromb Haemost ; 11(11): 2009-19, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24010820

RESUMEN

BACKGROUND: In vascular endothelial cells, high molecular weight multimers of von Willebrand factor (VWF) are folded into tubular structures for storage in Weibel-Palade bodies. On stimulation, VWF is secreted and forms strings to induce primary hemostasis. The structural changes composing the transition of stored tubular VWF into secreted unfurled VWF strings are still unresolved even though they are vital for normal hemostasis. The secretory pod is a novel structure that we previously described in endothelial cells. It is formed on stimulation and has been postulated to function as a VWF release site. In this study, we investigated the actual formation of secretory pods and the subsequent remodeling of VWF into strings. METHODS: Human umbilical vein endothelial cells were stimulated and studied using various imaging techniques such as live-cell imaging and correlative light and electron microscopy. RESULTS: We found by using live-cell imaging that secretory pods are formed through the coalescence of multiple Weibel-Palade bodies without involvement of other large structures. Secreted VWF expelled from secretory pods was found to adopt a globular conformation. We visualized that VWF strings derive from those globular masses of VWF. Flow experiments showed that, on secretion, the globular masses of VWF move to the edge of the cell, where they anchor and generate VWF strings. CONCLUSION: On secretion, VWF adopts a globular conformation that remodels into strings after translocation and anchoring at the edge of the cell. This finding reveals new pathophysiological mechanisms that could be affected in patients with von Willebrand disease.


Asunto(s)
Células Endoteliales/citología , Exocitosis , Factor de von Willebrand/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Hemostasis , Células Endoteliales de la Vena Umbilical Humana , Humanos , Fenotipo , Complejo GPIb-IX de Glicoproteína Plaquetaria/química , Unión Proteica , Conformación Proteica , Transporte de Proteínas , Cuerpos de Weibel-Palade/metabolismo
12.
Hamostaseologie ; 33(2): 121-30, 2013 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-23443151

RESUMEN

The von Willebrand factor (VWF)-cleaving metalloprotease, ADAMTS13 (adisintegrin and metalloprotease with thrombospondin type 1 motifs-13) is the only known target of the dysregulated immune response in acquired TTP. Autoantibodies to ADAMTS13 either neutralize its activity or accelerate its clearance, thereby causing a severe deficiency of ADAMTS13 in plasma. As a consequence, size regulation of VWF is impaired and the persistence of ultra-large VWF (ULVWF) multimers facilitates microvascular platelet aggregation causing microangiopathic haemolytic anaemia and ischaemic organ damage. Autoimmune TTP although a rare disease with an annual incidence of 1.72 cases has a mortality rate of 20% even with adequate therapy. We describe the mechanisms involved in ADAMTS13 autoimmunity with a focus on the role of B- and T-cells in the pathogenesis of this disorder. We discuss the potential translation of recent experimental findings into future therapeutic concepts for the treatment of acquired TTP.


Asunto(s)
Proteínas ADAM/inmunología , Autoinmunidad/inmunología , Linfocitos B/inmunología , Púrpura Trombocitopénica Trombótica/inmunología , Linfocitos T/inmunología , Factor de von Willebrand/inmunología , Proteína ADAMTS13 , Humanos , Modelos Inmunológicos , Púrpura Trombocitopénica Trombótica/patología
13.
J Thromb Haemost ; 11(1): 47-55, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23140211

RESUMEN

Only a fraction of patients with hemophilia A develop a neutralizing antibody (inhibitor) response to therapeutic infusions of factor VIII. Our present understanding of the underlying causes of the immunogenicity of this protein is limited. In the past few years, insights into the uptake and processing of FVIII by antigen-presenting cells (APCs) have expanded significantly. Although the mechanism of endocytosis remains unclear, current data indicate that FVIII enters APCs via its C1 domain. Its subsequent processing within endolysosomes allows for presentation of a heterogeneous collection of FVIII-derived peptides on major histocompatibility complex (MHC) class II, and this peptide-MHC class II complex may then be recognized by cognate effector CD4(+) T cells, leading to anti-FVIII antibody production. Here we aim to summarize recent knowledge gained about FVIII processing and presentation by APCs, as well as the diversity of the FVIII-specific T-cell repertoire in mice and humans. Moreover, we discuss possible factors that can drive FVIII immunogenicity. We believe that increasing understanding of the immune recognition of FVIII and the cellular mechanisms of anti-FVIII antibody production will lead to novel therapeutic approaches to prevent inhibitor formation in patients with hemophilia A.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Células Presentadoras de Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Coagulantes/inmunología , Factor VIII/inmunología , Hemofilia A/tratamiento farmacológico , Animales , Coagulantes/administración & dosificación , Coagulantes/química , Coagulantes/metabolismo , Endocitosis , Epítopos , Factor VIII/administración & dosificación , Factor VIII/química , Factor VIII/metabolismo , Hemofilia A/sangre , Hemofilia A/inmunología , Humanos , Activación de Linfocitos , Modelos Moleculares , Conformación Proteica , Factor de von Willebrand/metabolismo
14.
J Thromb Haemost ; 10(10): 2168-78, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22905953

RESUMEN

BACKGROUND AND OBJECTIVE: Von Willebrand factor (VWF) forms strings on activated vascular endothelial cells that recruit platelets and initiate clot formation. Alterations in VWF strings may disturb hemostasis. This study was aimed at developing a flexible model system for structure-function studies of VWF strings. METHODS: VWF strings were generated by inducing exocytosis of pseudo-Weibel-Palade bodies from VWF-transfected HEK293 cells, and the properties of these strings under static conditions and under flow were characterized. RESULTS: Upon exocytosis, VWF unfurled into strings several hundred micrometers in length. These strings could form bundles and networks, and bound platelets under flow, resembling authentic endothelial VWF strings. Anchorage of the platelet-decorated VWF strings was independent of P-selectin and integrin α(V) ß(3). Translocation of platelets along the strings, elongation and fragmentation of the strings frequently occurred under flow. Furthermore, VWF variants with the p.Tyr87Ser and p.Cys2773Ser mutations, which are defective in multimer assembly, did not give rise to VWF strings. Also, insertion of the green fluorescent protein into VWF inhibited string formation. CONCLUSIONS: HEK293 cells provide a flexible and useful model system for the study of VWF string formation. Our results suggest that structural changes in VWF may modulate string formation and function, and contribute to hemostatic disorders.


Asunto(s)
Plaquetas/metabolismo , Adhesividad Plaquetaria , Factor de von Willebrand/metabolismo , Exocitosis , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Integrina alfaVbeta3/metabolismo , Mutación , Selectina-P/metabolismo , Conformación Proteica , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/metabolismo , Relación Estructura-Actividad , Factores de Tiempo , Transfección , Cuerpos de Weibel-Palade/metabolismo , Factor de von Willebrand/química , Factor de von Willebrand/genética
15.
J Thromb Haemost ; 9(7): 1285-91, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21535387

RESUMEN

The apparently spontaneous development of autoantibodies to ADAMTS13 in previously healthy individuals is a major cause of thrombotic thrombocytopenic purpura (TTP). Epitope mapping studies have shown that in most patients antibodies directed towards the spacer domain of ADAMTS13 are present. A single antigenic surface comprising Arg(660) , Tyr(661) and Tyr(665) that contributes to the productive binding of ADAMTS13 to unfolded von Willebrand factor is targeted by anti-spacer domain antibodies. Antibodies directed to the carboxyl-terminal CUB1-2 and TSP2-8 domains have also been observed in the plasma of patients with acquired TTP. As yet it has not been established whether this class of antibodies modulates ADAMTS13 activity. Inspection of the primary sequence of human monoclonal anti-ADAMTS13 antibodies suggests that the variable heavy chain germline gene segment VH1-69 is frequently incorporated. We suggest a model in which 'shape complementarity' between the spacer domain and residues encoded by the VH1-69 gene segment explain the preferential use of this variable heavy chain gene segment. Finally, a model is presented for the development of anti-ADAMTS13 antibodies in previously healthy individuals that incorporates the recent identification of HLA DRB1*11 as a risk factor for acquired TTP.


Asunto(s)
Proteínas ADAM/inmunología , Inmunidad Humoral , Púrpura Trombocitopénica Trombótica/inmunología , Proteína ADAMTS13 , Autoanticuerpos/sangre , Mapeo Epitopo , Cadenas HLA-DRB1 , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/genética , Estructura Terciaria de Proteína , Factores de Riesgo
16.
J Thromb Haemost ; 9(4): 738-47, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21255251

RESUMEN

BACKGROUND: Patients with antiphospholipid syndrome (APS) display a heterogeneous population of antibodies with beta(2) glycoprotein-1 (ß(2)GP1) as the major antigen. OBJECTIVES: We isolated and characterized human mAbs directed against ß(2)GP1 from the immune repertoire of APS patients. METHODS: Variable heavy chain repertoires from B cells from two APS patients with anti-ß(2)GP1 antibodies were cloned into the pHEN1-VLrep vector. Constructed full-length IgG antibodies were tested for lupus anticoagulant (LAC) activity and binding to ß(2)GP1 and its domains. RESULTS: Two clones of each patient were selected on the basis of the reactivity of single chain Fv (scFv) fragments displayed on phages towards full-length ß(2)GP1 and its isolated domain I. The affinity of selected antibodies for ß(2)GP1 was lost when transforming from phages to monovalent scFvs, and was regained when antibodies were constructed as complete IgG, indicating a role for bivalency in binding to ß(2)GP1. Both selected clones from patient 2 recognized domain I of ß(2)GP1, and for both clones selected from patient 1, binding required the presence of both domain I and domain II. All mAbs displayed LAC activity in both activated partial thromboplastin time-based and dilute Russell's viper venom test-based clotting assays and in thrombin generation. CONCLUSIONS: In this study, we show successful cloning of patient-derived mAbs that require domain I of ß(2)GP1 for binding, and that display LAC activity that is dependent on their affinity for ß(2)GP1. These antibodies can help us to gain more insights into the pathogenesis of APS, and may facilitate standardization of APS diagnosis.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Inhibidor de Coagulación del Lupus/uso terapéutico , beta 2 Glicoproteína I/inmunología , beta 2 Glicoproteína I/uso terapéutico , Secuencia de Aminoácidos , Anticuerpos Monoclonales/química , Ensayo de Inmunoadsorción Enzimática , Humanos , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido
17.
J Thromb Haemost ; 9(4): 689-99, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21251204

RESUMEN

BACKGROUND: Development of neutralizing anti-factor (F)VIII antibodies ('inhibitors') is a serious clinical problem in hemophilia A. Increased inhibitor risk has been associated with certain FVIII missense substitutions, including R593C in the A2 domain. OBJECTIVES: The aim of the present study was to identify T-cell epitopes in FVIII and characterize T-cell responses in two unrelated hemophilia A subjects sharing F8-R593C and HLA-DRB1*1101 genotypes. We hypothesized that the hemophilic substitution site coincides with an important T-cell epitope. PATIENTS/METHODS: The binding affinities of peptides for recombinant HLA-DR proteins were measured and compared with epitope prediction results. CD4+ T cells were stimulated using peptides and stained with fluorescent, peptide-loaded tetramers. RESULTS: The inhibitor subjects, but not HLA-matched controls, had high-avidity HLA-DRB1*1101-restricted T-cell responses against FVIII(589-608), which contains the hemophilic missense site. Antigen-specific T cells secreted Th1 and Th2 cytokines and proliferated in response to FVIII and FVIII(592-603). FVIII(589-608) bound with physiologically relevant (micromolar) IC(50) values to recombinant DR0101, DR1101 and DR1501 proteins. CONCLUSIONS: Hemophilia A patients with R593C missense substitutions and these HLA haplotypes had an increased incidence of inhibitors in our cohorts, supporting a paradigm in which presentation of FVIII epitopes containing the wild-type R593 influences inhibitor risk in this hemophilia A sub-population.


Asunto(s)
Epítopos/inmunología , Factor VIII/genética , Hemofilia A/inmunología , Mutación Missense , Linfocitos T/inmunología , Secuencia de Aminoácidos , Proliferación Celular , Ensayo de Inmunoadsorción Enzimática , Antígenos HLA-DR/genética , Cadenas HLA-DRB1 , Humanos , Datos de Secuencia Molecular , Linfocitos T/citología
18.
Haemophilia ; 16(102): 35-43, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20536984

RESUMEN

Development of inhibitory antibodies to factor VIII (FVIII) provides a major complication of replacement therapy in patients with haemophilia A. The risk of inhibitor formation is influenced by the underlying FVIII gene defect. Moreover, genetic determinants in the promoter region of IL-10 and TNFalpha have been linked to an increased risk of inhibitor development. Recent cohort-studies have provided evidence that the risk of inhibitor formation is linked to intensity of treatment. Eradication of FVIII inhibitors can be achieved by frequent infusion of high dosages of FVIII, so-called immune tolerance induction (ITI). Until now, the mechanisms involved in downmodulation of the immune response to FVIII during ITI have not been unraveled. Studies performed in an animal model for haemophilia A have suggested that elimination of FVIII-specific memory B cells by high dosages of FVIII contributes to the decline in FVIII inhibitor levels during ITI. Limited knowledge is available with respect to the development and persistence of FVIII-specific memory B cells in patients with haemophilia A. Two recent studies suggest that the frequency of peripheral FVIII-specific memory B cells in haemophilia A patients with inhibitors range from <0.01 to 0.40% of that of total IgG(+) B cells. No or very low frequencies of FVIII-specific memory B cells are observed in haemophilia A patients without inhibitors and in patients treated successfully by ITI. Possible implications of these findings are discussed in the context of currently available information on the role of antigen-specific memory B cells and long-living antibody producing plasma cells in humoral immunity.


Asunto(s)
Linfocitos B/inmunología , Factor VIII/inmunología , Hemofilia A/inmunología , Tolerancia Inmunológica , Memoria Inmunológica/inmunología , Inhibidores de Factor de Coagulación Sanguínea/metabolismo , Ensayo de Inmunoadsorción Enzimática , Factor VIII/uso terapéutico , Hemofilia A/terapia , Humanos , Factores de Riesgo
19.
Haemophilia ; 16(6): 892-901, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20491952

RESUMEN

INTRODUCTION: Frequent administration of high dosages factor VIII (FVIII), so-called immune tolerance induction (ITI), provides an efficient strategy to eradicate inhibitory antibodies in patients with haemophilia A. At present, our knowledge on the characteristics of inhibitory antibodies in patients undergoing ITI is limited. AIM: In this study we characterized the domain specificity of FVIII inhibitors in 11 haemophilia A patients during ITI. RESULTS: In three of six patients who were successfully tolerized, inhibitory antibodies were directed predominantly against the FVIII light chain. In two other patients within this group, a significant contribution of A2 antibodies was observed which did not change during treatment. In the sixth patient the relative contribution of A2 inhibitors declined which coincided with an increase in antilight chain antibodies. In four of five patients who failed ITI, A2 inhibitors were observed. In two patients the contribution of A2 inhibitors increased during treatment, while in two other patients the contribution of A2 inhibitor remained constant. The fifth patient had inhibitory antibodies predominantly directed against the FVIII light chain. CONCLUSION: Overall, our findings revealed changes in domain specificity of FVIII antibodies in five of 11 patients analysed. Remarkably, antibodies exclusively directed towards the light chain of FVIII were predominantly observed in patients who were successfully tolerized.


Asunto(s)
Inhibidores de Factor de Coagulación Sanguínea/inmunología , Factor VIII/inmunología , Hemofilia A/inmunología , Tolerancia Inmunológica , Especificidad de Anticuerpos , Estudios de Cohortes , Factor VIII/administración & dosificación , Factor VIII/genética , Hemofilia A/complicaciones , Hemofilia A/tratamiento farmacológico , Humanos , Mutación
20.
Haemophilia ; 15(3): 712-7, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19432924

RESUMEN

Both genetic factors and environmental factors are suggested to play a role in the aetiology of inhibitor development in patients with severe haemophilia A. Monozygotic twins are ideal candidates to study the influence of environmental factors. We describe a pair of 3-year-old monozygotic twin brothers with severe haemophilia A. Prenatal diagnosis confirmed the presence of an intron 22 inversion. Other patient-related factors such as birth weight, vaccinations and the duration of breastfeeding were similar. At the age of 7 months, one boy suffered from a spontaneous subdural haematoma, which needed complete correction of haemostasis with continuous infusion of a third-generation recombinant factor VIII. A persistent high-titre inhibitor with severe clinical symptoms developed, that could only be eradicated with high-dose immune tolerance induction (ITI) for 36 months in combination with rituximab therapy. His twin brother first received treatment at 9 months of age with the same FVIII product. After treatment on nine exposure days, he developed a low-titre inhibitor at the age of 14.5 months. Unlike his brother, he was tolerized without difficulties with low-dose ITI within 2 months. The discordant antibody responses were underlined by dissimilar IgG1 and IgG4 levels in their plasma. The discordant immune response to FVIII in this pair of monozygotic twin brothers seemed to be related to intensity of treatment and severity of bleeds. This confirms that these environmental factors play an additional role in the development of inhibitors.


Asunto(s)
Inhibidores de Factor de Coagulación Sanguínea/inmunología , Factor VIII/uso terapéutico , Hemartrosis/inmunología , Hematoma Intracraneal Subdural/diagnóstico , Hemofilia A/inmunología , Preescolar , Factor VIII/efectos adversos , Factor VIII/inmunología , Hemartrosis/complicaciones , Hemartrosis/tratamiento farmacológico , Hemofilia A/complicaciones , Hemofilia A/tratamiento farmacológico , Humanos , Masculino , Índice de Severidad de la Enfermedad , Gemelos Monocigóticos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...