Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Thromb Haemost ; 18(6): 1278-1290, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32108991

RESUMEN

BACKGROUND: Previous studies have demonstrated that the A1A2A3 domains of von Willebrand factor (VWF) play a key role in regulating macrophage-mediated clearance in vivo. In particular, the A1-domain has been shown to modulate interaction with macrophage low-density lipoprotein receptor-related protein-1 (LRP1) clearance receptor. Furthermore, N-linked glycans within the A2-domain have been shown to protect VWF against premature LRP1-mediated clearance. Importantly, however, the specific regions within A1A2A3 that enable macrophage binding have not been defined. OBJECTIVE AND METHODS: To address this, we utilized site-directed PEGylation and introduced novel targeted N-linked glycosylation within A1A2A3-VWF and subsequently examined VWF clearance. RESULTS: Conjugation with a 40-kDa polyethylene glycol (PEG) moiety significantly extended the half-life of A1A2A3-VWF in VWF-/- mice in a site-specific manner. For example, PEGylation at specific sites within the A1-domain (S1286) and A3-domain (V1803, S1807) attenuated VWF clearance in vivo, compared to wild-type A1A2A3-VWF. Furthermore, PEGylation at these specific sites ablated binding to differentiated THP-1 macrophages and LRP1 cluster II and cluster IV in-vitro. Conversely, PEGylation at other positions (Q1353-A1-domain and M1545-A2-domain) had limited effects on VWF clearance or binding to LRP1.Novel N-linked glycan chains were introduced at N1803 and N1807 in the A3-domain. In contrast to PEGylation at these sites, no significant extension in half-life was observed with these N-glycan variants. CONCLUSIONS: These novel data demonstrate that site specific PEGylation but not site specific N-glycosylation modifies LRP1-dependent uptake of the A1A2A3-VWF by macrophages. This suggests that PEGylation, within the A1- and A3-domains in particular, may be used to attenuate LRP1-mediated clearance of VWF.


Asunto(s)
Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Factor de von Willebrand , Animales , Glicosilación , Cinética , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Ratones , Polisacáridos , Unión Proteica , Factor de von Willebrand/metabolismo
2.
Blood ; 132(14): 1473-1477, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30097509

RESUMEN

Decreased hepcidin mobilizes iron, which facilitates erythropoiesis, but excess iron is pathogenic in ß-thalassemia. Erythropoietin (EPO) enhances erythroferrone (ERFE) synthesis by erythroblasts, and ERFE suppresses hepatic hepcidin production through an unknown mechanism. The BMP/SMAD pathway in the liver is critical for hepcidin control, and we show that EPO suppressed hepcidin and other BMP target genes in vivo in a partially ERFE-dependent manner. Furthermore, recombinant ERFE suppressed the hepatic BMP/SMAD pathway independently of changes in serum and liver iron. In vitro, ERFE decreased SMAD1, SMAD5, and SMAD8 phosphorylation and inhibited expression of BMP target genes. ERFE specifically abrogated the induction of hepcidin by BMP5, BMP6, and BMP7 but had little or no effect on hepcidin induction by BMP2, BMP4, BMP9, or activin B. A neutralizing anti-ERFE antibody prevented ERFE from inhibiting hepcidin induction by BMP5, BMP6, and BMP7. Cell-free homogeneous time-resolved fluorescence assays showed that BMP5, BMP6, and BMP7 competed with anti-ERFE for binding to ERFE. We conclude that ERFE suppresses hepcidin by inhibiting hepatic BMP/SMAD signaling via preferentially impairing an evolutionarily closely related BMP subgroup of BMP5, BMP6, and BMP7. ERFE can act as a natural ligand trap generated by stimulated erythropoiesis to regulate the availability of iron.


Asunto(s)
Proteína Morfogenética Ósea 6/metabolismo , Citocinas/metabolismo , Hepcidinas/metabolismo , Proteínas Musculares/metabolismo , Animales , Línea Celular , Células Hep G2 , Humanos , Hierro/metabolismo , Hígado/metabolismo , Masculino , Ratones , Transducción de Señal , Proteínas Smad/metabolismo
3.
J Biol Chem ; 291(3): 1267-76, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26515064

RESUMEN

Fully-human single-chain Fv (scFv) proteins are key potential building blocks of bispecific therapeutic antibodies, but they often suffer from manufacturability and clinical development limitations such as instability and aggregation. The causes of these scFv instability problems, in proteins that should be theoretically stable, remains poorly understood. To inform the future development of such molecules, we carried out a comprehensive structural analysis of the highly stabilized anti-CXCL13 scFv E10. E10 was derived from the parental 3B4 using complementarity-determining region (CDR)-restricted mutagenesis and tailored selection and screening strategies, and carries four mutations in VL-CDR3. High-resolution crystal structures of parental 3B4 and optimized E10 scFvs were solved in the presence and absence of human CXCL13. In parallel, a series of scFv mutants was generated to interrogate the individual contribution of each of the four mutations to stability and affinity improvements. In combination, these analyses demonstrated that the optimization of E10 was primarily mediated by removing clashes between both the VL and the VH, and between the VL and CXCL13. Importantly, a single, germline-encoded VL-CDR3 residue mediated the key difference between the stable and unstable forms of the scFv. This work demonstrates that, aside from being the critical mediators of specificity and affinity, CDRs may also be the primary drivers of biotherapeutic developability.


Asunto(s)
Productos Biológicos/química , Quimiocina CXCL13/antagonistas & inhibidores , Modelos Moleculares , Anticuerpos de Cadena Única/química , Sustitución de Aminoácidos , Afinidad de Anticuerpos , Especificidad de Anticuerpos , Complejo Antígeno-Anticuerpo/química , Complejo Antígeno-Anticuerpo/metabolismo , Sitios de Unión de Anticuerpos , Productos Biológicos/metabolismo , Quimiocina CXCL13/química , Quimiocina CXCL13/metabolismo , Regiones Determinantes de Complementariedad/química , Regiones Determinantes de Complementariedad/genética , Regiones Determinantes de Complementariedad/metabolismo , Humanos , Cinética , Mutación , Agregado de Proteínas , Conformación Proteica , Estabilidad Proteica , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/metabolismo , Solubilidad , Difracción de Rayos X
4.
Blood ; 115(13): 2666-73, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19965639

RESUMEN

von Willebrand factor (VWF) multimeric composition is regulated in plasma by ADAMTS13. VWF deglycosylation enhances proteolysis by ADAMTS13. In this study, the role of terminal sialic acid residues on VWF glycans in mediating proteolysis by ADAMTS13 was investigated. Quantification and distribution of VWF sialylation was examined by sequential digestion and high-performance liquid chromatography analysis. Total sialic acid expression on VWF was 167nmol/mg, of which the majority (80.1%) was present on N-linked glycan chains. Enzymatic desialylation of VWF by alpha2-3,6,8,9 neuraminidase (Neu-VWF) markedly impaired ADAMTS13-mediated VWF proteolysis. Neu-VWF collagen binding activity was reduced to 50% (+/- 14%) by ADAMTS13, compared with 11% (+/- 7%) for untreated VWF. Despite this, Neu-VWF exhibited increased susceptibility to other proteases, including trypsin, chymotrypsin, and cathepsin B. VWF expressing different blood groups exhibit altered ADAMTS13 proteolysis rates (O > or = B > A > or = AB). However, ABO blood group regulation of ADAMTS13 proteolysis was ablated on VWF desialylation, as both Neu-O-VWF and Neu-AB-VWF were cleaved by ADAMTS13 at identical rates. These novel data show that sialic acid protects VWF against proteolysis by serine and cysteine proteases but specifically enhances susceptibility to ADAMTS13 proteolysis. Quantitative variation in VWF sialylation therefore represents a key determinant of VWF multimeric composition and, as such, may be of pathophysiologic significance.


Asunto(s)
Proteínas ADAM/metabolismo , Ácido N-Acetilneuramínico/fisiología , Factor de von Willebrand/química , Sistema del Grupo Sanguíneo ABO/química , Sistema del Grupo Sanguíneo ABO/metabolismo , Proteína ADAMTS13 , Biopolímeros , Conformación de Carbohidratos , Colágeno/metabolismo , Proteasas de Cisteína/metabolismo , Galactosa/química , Glicósido Hidrolasas/farmacología , Humanos , Ácido N-Acetilneuramínico/química , Neuraminidasa/farmacología , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/farmacología , Conformación Proteica , Procesamiento Proteico-Postraduccional , Serina Proteasas/metabolismo , Especificidad por Sustrato , alfa-N-Acetilgalactosaminidasa/farmacología , Factor de von Willebrand/efectos de los fármacos , Factor de von Willebrand/metabolismo
5.
PLoS Pathog ; 5(3): e1000349, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19300493

RESUMEN

Plasmodium falciparum infection results in adhesion of infected erythrocytes to blood vessel endothelium, and acute endothelial cell activation, together with sequestration of platelets and leucocytes. We have previously shown that patients with severe infection or fulminant cerebral malaria have significantly increased circulatory levels of the adhesive glycoprotein von Willebrand factor (VWF) and its propeptide, both of which are indices of endothelial cell activation. In this prospective study of patients from Ghana with severe (n = 20) and cerebral (n = 13) P. falciparum malaria, we demonstrate that increased plasma VWF antigen (VWF:Ag) level is associated with disproportionately increased VWF function. VWF collagen binding (VWF:CB) was significantly increased in patients with cerebral malaria and severe malaria (medians 7.6 and 7.0 IU/ml versus 1.9 IU/ml; p<0.005). This increased VWF:CB correlated with the presence of abnormal ultra-large VWF multimers in patient rather than control plasmas. Concomitant with the increase in VWF:Ag and VWF:CB was a significant persistent reduction in the activity of the VWF-specific cleaving protease ADAMTS13 (approximately 55% of normal; p<0.005). Mixing studies were performed using P. falciparum patient plasma and normal pooled plasma, in the presence or absence of exogenous recombinant ADAMTS13. These studies demonstrated that in malarial plasma, ADAMTS13 function was persistently inhibited in a time-dependent manner. Furthermore, this inhibitory effect was not associated with the presence of known inhibitors of ADAMTS13 enzymatic function (interleukin-6, free haemoglobin, factor VIII or thrombospondin-1). These novel findings suggest that severe P. falciparum infection is associated with acute endothelial cell activation, abnormal circulating ULVWF multimers, and a significant reduction in plasma ADAMTS13 function which is mediated at least in part by an unidentified inhibitor.


Asunto(s)
Proteínas ADAM/metabolismo , Células Endoteliales/metabolismo , Malaria Falciparum/metabolismo , Factor de von Willebrand/metabolismo , Proteína ADAMTS13 , Niño , Preescolar , Ensayo de Inmunoadsorción Enzimática , Humanos , Lactante , Multimerización de Proteína
6.
Thromb Res ; 120 Suppl 2: S64-70, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18023715

RESUMEN

Von Willebrand factor (VWF) is a multimeric plasma glycoprotein that plays a critical role in primary hemostasis, allowing the adhesion of platelets to the exposed subendothelium. The key role played by VWF in platelet adhesion suggests a potential implication in various pathologies where this process is involved. In cancer metastasis development, tumor cells interact with platelets and the vessel wall to extravasate from the circulation. A number of potential receptors for VWF have been identified on tumor cells such as glycoprotein Ib or the alpha(IIb)beta(3) and alpha(v)beta(3) integrins and direct interactions between VWF and tumor cells have been reported. To address the role of VWF in an experimental metastasis model, we compared the formation of pulmonary metastatic foci in C57BL/6J wild-type and VWF-null mice following I.V. injection of murine melanoma B16-BL6 cells or Lewis lung carcinoma cells. Surprisingly we found a significant increase in the number of pulmonary metastatic foci in VWF-null mice. Restoration of VWF plasma levels by co-injection of VWF with the tumor cells led to the correction of this pro-metastatic phenotype. In vitro analysis revealed that VWF did not influence tumor cell proliferation or invasion but induced cellular death. This result was confirmed in vivo where analysis of the early survival of tumor cells in the lungs revealed that the presence of VWF led to a decreased survival of these cells during the first 24 hours after injection. Our results suggest that VWF plays a role in tumor metastasis, independently of its role in hemostasis.


Asunto(s)
Metástasis de la Neoplasia , Factor de von Willebrand/fisiología , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Modelos Animales de Enfermedad , Neoplasias Pulmonares/secundario , Melanoma/secundario , Ratones , Ratones Endogámicos C57BL , Invasividad Neoplásica , Adhesividad Plaquetaria , Neoplasias Cutáneas/patología
7.
Blood ; 108(12): 3746-52, 2006 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16926295

RESUMEN

Von Willebrand factor (VWF) is an essential component of hemostasis. However, animal studies using VWF-deficient mice suggest that VWF may also contribute to inflammation. In the present study, we demonstrate that VWF was able to interact with polymorphonuclear leukocytes (PMNs) and monocytes under static and flow conditions. Adhesion under flow was dominated by short-lasting contact with resting PMNs, whereas adhesion of phorbol-12-myristate-13-acetate (PMA)-stimulated PMNs was characterized by firm adhesion. Transient binding of PMNs to VWF appeared to be mediated by P-selectin glycoprotein ligand-1 (PSGL-1). Moreover, recombinant PSGL-1 protein and cell surface-expressed PSGL-1 directly interacted with VWF. As for stable adhesion by PMA-stimulated PMNs, we observed that static adhesion and adhesion under flow were strongly inhibited (greater than 75%) by neutrophil-inhibitory factor, an inhibitor of beta2-integrin function. In addition, the isolated I-domain of alphaMbeta2 bound to VWF, and cell lines expressing alphaLbeta2 or alphaXbeta2 adhered efficiently to VWF. Taken together, our data showed that VWF can function as an adhesive surface for various leukocyte subsets (monocytes, PMNs). Analogous to VWF-platelet interaction, VWF provided binding sites for leukocyte receptors involved in rolling (PSGL-1) and stable (beta2-integrins) adhesion. VWF is unique in its intrinsic capacity to combine the rolling and the stable adhesion step in the interaction with leukocytes.


Asunto(s)
Antígenos CD18/metabolismo , Granulocitos/metabolismo , Rodamiento de Leucocito/fisiología , Glicoproteínas de Membrana/metabolismo , Monocitos/metabolismo , Factor de von Willebrand/metabolismo , Animales , Sitios de Unión/fisiología , Plaquetas/citología , Plaquetas/metabolismo , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Granulocitos/citología , Humanos , Rodamiento de Leucocito/efectos de los fármacos , Glicoproteínas de Membrana/farmacología , Ratones , Monocitos/citología , Estructura Terciaria de Proteína , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Células U937 , Enfermedades de von Willebrand/metabolismo
8.
J Biol Chem ; 279(13): 12102-9, 2004 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-14613933

RESUMEN

To explore the molecular basis of von Willebrand factor (VWF) clearance, an experimental model employing VWF-deficient mice was developed. Biodistribution was examined by the injection of radiolabeled VWF, which was primarily directed to the liver with minor amounts in other organs. Disappearance of VWF from plasma was characterized by a rapid initial phase (t((1/2))alpha = 13 min) and a slow secondary phase (t((1/2))beta = 3 h), with a mean residence time (MRT) of 2.8 h. A similar clearance was observed for VWF consisting of only high or low molecular weight multimers, indicating that, in our experimental model, clearance is independent of multimeric distribution. This allowed us to compare the survival of full-length VWF to truncated variants. Deletion of both the amino-terminal D'-D3 and carboxyl-terminal D4-CK domains resulted in a fragment with a similar clearance to wild-type VWF. Deletion of only the D'-D3 region was associated with an almost 2-fold lower recovery and increased clearance (MRT = 1.6 h), whereas deletion of only the D4-CK region resulted in a significantly reduced clearance (MRT = 4.5 h, p < 0.02). These results point to a role of the D'-D3 region in preventing clearance of VWF. Furthermore, replacement of D3 domain residue Arg-1205 by His resulted in a markedly increased clearance (MRT = 0.3 h; p = 0.004). Therefore, this mutation seems to abrogate the protective effect of the D'-D3 region. In vitro analysis of this mutant also revealed a 2-fold reduced affinity for VWF propeptide at low pH, showing that mutation of Arg-1205 results not only in an increased clearance rate but is also associated with an impaired pH-dependent interaction with VWF propeptide.


Asunto(s)
Mutación , Glicoproteínas de Membrana Plaquetaria , Factor de von Willebrand/química , Animales , Arginina/química , Línea Celular , Colágeno/química , Cricetinae , Eliminación de Gen , Histidina/química , Concentración de Iones de Hidrógeno , Cinética , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Péptidos/química , Complejo GPIb-IX de Glicoproteína Plaquetaria/química , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Resonancia por Plasmón de Superficie , Factores de Tiempo , Distribución Tisular , Factor de von Willebrand/genética , Factor de von Willebrand/farmacocinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...