Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Haematologica ; 102(4): 686-694, 2017 04.
Article in English | MEDLINE | ID: mdl-28057741

ABSTRACT

Factor VIII C-domains are believed to have specific functions in cofactor activity and in interactions with von Willebrand factor. We have previously shown that factor VIII is co-targeted with von Willebrand factor to the Weibel-Palade bodies in blood outgrowth endothelial cells, even when factor VIII carries mutations in the light chain that are associated with defective von Willebrand factor binding. In this study, we addressed the contribution of individual factor VIII C-domains in intracellular targeting, von Willebrand factor binding and cofactor activity by factor VIII/V C-domain swapping. Blood outgrowth endothelial cells were transduced with lentivirus encoding factor V, factor VIII or YFP-tagged C-domain chimeras, and examined by confocal microscopy. The same chimeras were produced in HEK293-cells for in vitro characterization and chemical foot-printing by mass spectrometry. In contrast to factor VIII, factor V did not target to Weibel-Palade bodies. The chimeras showed reduced Weibel-Palade body targeting, suggesting that this requires the factor VIII C1-C2 region. The factor VIII/V-C1 chimera did not bind von Willebrand factor and had reduced affinity for activated factor IX, whereas the factor VIII/V-C2 chimera showed a minor reduction in von Willebrand factor binding and normal interaction with activated factor IX. This suggests that mainly the C1-domain carries factor VIII-specific features in assembly with von Willebrand factor and activated factor IX. Foot-printing analysis of the chimeras revealed increased exposure of lysine residues in the A1/C2- and C1/C2-domain interface, suggesting increased C2-domain mobility and disruption of the natural C-domain tandem pair orientation. Apparently, this affects intracellular trafficking, but not extracellular function.


Subject(s)
Factor VIII/metabolism , Factor V/metabolism , Protein Interaction Domains and Motifs , Endothelial Cells/metabolism , Factor V/chemistry , Factor V/genetics , Factor VIII/chemistry , Factor VIII/genetics , Gene Expression , Humans , Intracellular Space/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Protein Transport , Structure-Activity Relationship , von Willebrand Factor/metabolism
2.
Thromb Haemost ; 114(6): 1144-55, 2015 Nov 25.
Article in English | MEDLINE | ID: mdl-26272345

ABSTRACT

The protein C (PC) system conveys beneficial anticoagulant and cytoprotective effects in numerous in vivo disease models. The endothelial protein C receptor (EPCR) plays a central role in these pathways as cofactor for PC activation and by enhancing activated protein C (APC)-mediated protease-activated receptor (PAR) activation. During inflammatory disease, expression of EPCR on cell membranes is often diminished thereby limiting PC activation and APC's effects on cells. Here a caveolae-targeting glycosylphosphatidylinositol (GPI)-anchored EPCR (EPCR-GPI) was engineered to restore EPCR's bioavailability via "cell painting." The painting efficiency of EPCR-GPI on EPCR-depleted endothelial cells was time- and dose-dependent. The EPCR-GPI bioavailability after painting was long lasting since EPCR surface levels reached 400 % of wild-type cells after 2 hours and remained > 200 % for 24 hours. EPCR-GPI painting conveyed APC binding to EPCR-depleted endothelial cells where EPCR was lost due to shedding or shRNA. EPCR painting normalised PC activation on EPCR-depleted cells indicating that EPCR-GPI is functional active on painted cells. Caveolin-1 lipid rafts were enriched in EPCR after painting due to the GPI-anchor targeting caveolae. Accordingly, EPCR painting supported PAR1 and PAR3 cleavage by APC and augmented PAR1-dependent Akt phosphorylation by APC. Thus, EPCR-GPI painting achieved physiological relevant surface levels on endothelial cells, restored APC binding to EPCR-depleted cells, supported PC activation, and enhanced APC-mediated PAR cleavage and cytoprotective signalling. Therefore, EPCR-GPI provides a novel tool to restore the bioavailability and functionality of EPCR on EPCR- depleted and -deficient cells.


Subject(s)
Antigens, CD/metabolism , Caveolae/metabolism , Cell Membrane/metabolism , Endothelial Cells/metabolism , Glycosylphosphatidylinositols/metabolism , Membrane Microdomains/metabolism , Protein C/metabolism , Receptors, Cell Surface/metabolism , 3' Untranslated Regions/genetics , Antigens, CD/genetics , Biological Availability , Caveolin 1/metabolism , Cell Compartmentation , Dose-Response Relationship, Drug , Endothelial Protein C Receptor , Enzyme Activation , HEK293 Cells , Humans , Phosphorylation , Protein Binding , Protein Engineering , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering/genetics , Receptor, PAR-1/metabolism , Receptors, Cell Surface/genetics , Recombinant Fusion Proteins/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transduction, Genetic
3.
Thromb Haemost ; 114(5): 1038-48, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26155776

ABSTRACT

The Endothelial Protein C receptor (EPCR) is essential for the anticoagulant and cytoprotective functions of the Protein C (PC) system. Selected variants of the malaria parasite protein, Plasmodium falciparum Erythrocyte Membrane Protein 1 (PfEMP1) associated with severe malaria, including cerebral malaria, specifically target EPCR on vascular endothelial cells. Here, we examine the cellular response to PfEMP1 engagement to elucidate its role in malaria pathogenesis. Binding of the CIDRα1.1 domain of PfEMP1 to EPCR obstructed activated PC (APC) binding to EPCR and induced a loss of cellular EPCR functions. CIDRα1.1 severely impaired endothelial PC activation and effectively blocked APC-mediated activation of protease-activated receptor-1 (PAR1) and associated barrier protective effects of APC on endothelial cells. A soluble EPCR variant (E86A-sEPCR) bound CIDRα1.1 with high affinity and did not interfere with (A)PC binding to cellular EPCR. E86A-sEPCR used as a decoy to capture PfEMP1, permitted normal PC activation on endothelial cells, normal barrier protective effects of APC, and greatly reduced cytoadhesion of infected erythrocytes to brain endothelial cells. These data imply important contributions of PfEMP1-induced protein C pathway defects in the pathogenesis of severe malaria. Furthermore, the E86A-sEPCR decoy provides a proof-of-principle strategy for the development of novel adjunct therapies for severe malaria.


Subject(s)
Antigens, CD/metabolism , Endothelial Cells/physiology , Malaria/physiopathology , Plasmodium falciparum/physiology , Protozoan Proteins/metabolism , Receptors, Cell Surface/metabolism , Antigens, CD/genetics , Brain/pathology , Cells, Cultured , Cytoprotection/drug effects , Endothelial Cells/drug effects , Endothelial Cells/parasitology , Endothelial Protein C Receptor , Humans , Malaria/drug therapy , Malaria/parasitology , Mutant Proteins/administration & dosage , Protein Binding/drug effects , Protein C/metabolism , Protein Structure, Tertiary/genetics , Protozoan Proteins/genetics , Receptor, PAR-1/metabolism , Receptors, Cell Surface/genetics , Signal Transduction/drug effects
4.
Blood ; 121(14): 2762-72, 2013 Apr 04.
Article in English | MEDLINE | ID: mdl-23426949

ABSTRACT

Patients with von Willebrand disease (VWD) are often heterozygous for a missense mutation in the von Willebrand factor (VWF) gene. Investigating the pathogenic features of VWF mutations in cells directly derived from patients has been challenging. Here, we have used blood outgrowth endothelial cells (BOECs) isolated from human peripheral blood to analyze the storage and secretion of VWF. BOECs showed full endothelial characteristics and responded to Weibel-Palade body (WPB) secretagogues except desmopressin. We examined BOECs derived from a single subject heterozygous for a type 2N mutation (p.Arg854Gln) and from 4 patients with type 1 VWD who were, respectively, heterozygous for p.Ser1285Pro, p.Leu1307Pro, p.Tyr1584Cys, and p.Cys2693Tyr. Compared with normal BOECs, BOECs heterozygous for p.Ser1285Pro, p.Leu1307Pro, or p.Cys2693Tyr showed morphologically abnormal WPB and retention of VWF in the endoplasmic reticulum, whereas BOECs heterozygous for p.Arg854Gln or p.Tyr1584Cys showed normal WPB. The agonist-induced exocytosis of WPB from BOECs and formation of VWF strings on BOECs heterozygous for p.Ser1285Pro, p.Leu1307Pro, or p.Cys2693Tyr, but not for p.Arg854Gln or p.Tyr1584Cys, were reduced. In conclusion, VWD phenotype can be recapitulated in BOECs, and thus BOECs provide a feasible bona fide cell model to study the pathogenic effects of VWF mutations.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/metabolism , Weibel-Palade Bodies/metabolism , von Willebrand Disease, Type 1/metabolism , von Willebrand Factor/metabolism , Cells, Cultured , Endoplasmic Reticulum/metabolism , Endothelial Cells/physiology , Exocytosis/physiology , Female , Flow Cytometry , Genotype , Heterozygote , Humans , Male , Mutation, Missense , Phenotype , von Willebrand Disease, Type 1/genetics , von Willebrand Disease, Type 1/pathology , von Willebrand Factor/genetics
5.
Hematol Educ ; 7(1): 365-374, 2013.
Article in English | MEDLINE | ID: mdl-24741378

ABSTRACT

The protein C pathway provides important biological activities to maintain the fluidity of the circulation, prevent thrombosis, and protect the integrity of the vasculature in response to injury. Activated protein C (APC), in concert with its cofactors and cell receptors, assembles in specific macromolecular complexes to provide efficient proteolysis of multiple substrates that result in anticoagulant and cytoprotective activities. Numerous studies on APC's structure-function relation with its cofactors, cell receptors, and substrates provide valuable insights into the molecular mechanisms and presumed assembly of the macromolecular complexes that are responsible for APC's activities. These insights allow for molecular engineering approaches specifically targeting the interaction of APC with one of its substrates or cofactors. Thus far, these approaches resulted in several anticoagulant-selective and cytoprotective-selective APC mutants, which provide unique insights into the relative contributions of APC's anticoagulant or cytoprotective activities to the beneficial effects of APC in various murine injury and disease models. Because of its multiple physiological and pharmacological activities, the anticoagulant and cytoprotective protein C pathway have important implications for the (patho)physiology of vascular disease and for translational research exploring novel therapeutic strategies to combat complex medical disorders such as thrombosis, inflammation, ischemic stroke and neurodegenerative disease.

6.
PLoS One ; 6(8): e24163, 2011.
Article in English | MEDLINE | ID: mdl-21909383

ABSTRACT

BACKGROUND: Point mutations resulting in reduced factor VIII (FVIII) binding to von Willebrand factor (VWF) are an important cause of mild/moderate hemophilia A. Treatment includes desmopressin infusion, which concomitantly increases VWF and FVIII plasma levels, apparently from storage pools containing both proteins. The source of these VWF/FVIII co-storage pools and the mechanism of granule biogenesis are not fully understood. METHODOLOGY/PRINCIPAL FINDINGS: We studied intracellular trafficking of FVIII variants implicated in mild/moderate hemophilia A together with VWF in HEK293 cells and primary endothelial cells. The role of VWF binding was addressed using FVIII variants displaying reduced VWF interaction. Binding studies using purified FVIII proteins revealed moderate (Arg2150His, Del2201, Pro2300Ser) to severe (Tyr1680Phe, Ser2119Tyr) VWF binding defects. Expression studies in HEK293 cells and primary endothelial cells revealed that all FVIII variants were present within VWF-containing organelles. Quantitative studies showed that the relative amount of FVIII storage was independent of various mutations. Substantial amounts of FVIII variants are co-stored in VWF-containing storage organelles, presumably by virtue of their ability to interact with VWF at low pH. CONCLUSIONS: Our data suggest that the potential of FVIII co-storage with VWF is not affected in mild/moderate hemophilia A caused by reduced FVIII/VWF interaction in the circulation. These data support the hypothesis that Weibel-Palade bodies comprise the desmopressin-releasable FVIII storage pool in vivo.


Subject(s)
Endothelial Cells/metabolism , Factor VIII/metabolism , Mutant Proteins/metabolism , Weibel-Palade Bodies/metabolism , von Willebrand Factor/metabolism , Amino Acid Substitution , Cytoplasmic Granules/metabolism , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Intracellular Space/metabolism , Protein Binding , Protein Transport , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Subcellular Fractions/metabolism , Surface Plasmon Resonance , Transfection
7.
Blood ; 118(22): 5947-56, 2011 Nov 24.
Article in English | MEDLINE | ID: mdl-21940821

ABSTRACT

In endothelial cells, von Willebrand factor (VWF) multimers are packaged into tubules that direct biogenesis of elongated Weibel-Palade bodies (WPBs). WPB release results in unfurling of VWF tubules and assembly into strings that serve to recruit platelets. By confocal microscopy, we have previously observed a rounded morphology of WPBs in blood outgrowth endothelial cells transduced to express factor VIII (FVIII). Using correlative light-electron microscopy and tomography, we now demonstrate that FVIII-containing WPBs have disorganized, short VWF tubules. Whereas normal FVIII and FVIII Y1680F interfered with formation of ultra-large VWF multimers, release of the WPBs resulted in VWF strings of equal length as those from nontransduced blood outgrowth endothelial cells. After release, both WPB-derived FVIII and FVIII Y1680F remained bound to VWF strings, which however had largely lost their ability to recruit platelets. Strings from nontransduced cells, however, were capable of simultaneously recruiting exogenous FVIII and platelets. These findings suggest that the interaction of FVIII with VWF during WPB formation is independent of Y1680, is maintained after WPB release in FVIII-covered VWF strings, and impairs recruitment of platelets. Apparently, intra-cellular and extracellular assembly of FVIII-VWF complex involves distinct mechanisms, which differ with regard to their implications for platelet binding to released VWF strings.


Subject(s)
Factor VIII/pharmacology , Microtubules/metabolism , Protein Multimerization/drug effects , Weibel-Palade Bodies/metabolism , von Willebrand Factor/metabolism , von Willebrand Factor/physiology , Amino Acid Substitution , Blood Platelets/drug effects , Blood Platelets/metabolism , Blood Platelets/physiology , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/physiology , Factor VIII/genetics , Factor VIII/metabolism , Humans , Microtubules/drug effects , Microtubules/physiology , Phenylalanine/genetics , Protein Multimerization/genetics , Transfection , Tyrosine/genetics , Weibel-Palade Bodies/drug effects , Weibel-Palade Bodies/ultrastructure
8.
Haematologica ; 94(5): 670-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19336741

ABSTRACT

BACKGROUND: Gene therapy provides an attractive alternative for protein replacement therapy in hemophilia A patients. Recent studies have shown the potential benefit of directing factor (F)VIII gene delivery to cells that also express its natural carrier protein von Willebrand factor (VWF). In this study, we explored the feasibility of blood outgrowth endothelial cells as a cellular FVIII delivery device with particular reference to long-term production levels, intracellular storage in Weibel-Palade bodies and agonist-induced regulated secretion. DESIGN AND METHODS: Human blood outgrowth endothelial cells were isolated from peripheral blood collected from healthy donors, transduced at passage 5 using a lentiviral vector encoding human B-domain deleted FVIII-GFP and characterized by flow cytometry and confocal microscopy. RESULTS: Blood outgrowth endothelial cells displayed typical endothelial morphology and expressed the endothelial-specific marker VWF. Following transduction with a lentivirus encoding FVIII-GFP, 80% of transduced blood outgrowth endothelial cells expressed FVIII-GFP. Levels of FVIII-GFP positive cells declined slowly upon prolonged culturing. Transduced blood outgrowth endothelial cells expressed 1.6+/-1.0 pmol/1 x 10(6) cells/24h FVIII. Morphological analysis demonstrated that FVIII-GFP was stored in Weibel-Palade bodies together with VWF and P-selectin. FVIII levels were only slightly increased following agonist-induced stimulation, whereas a 6- to 8-fold increase of VWF levels was observed. Subcellular fractionation revealed that 15-22% of FVIII antigen was present within the dense fraction containing Weibel-Palade bodies. CONCLUSIONS: We conclude that blood outgrowth endothelial cells, by virtue of their ability to store a significant portion of synthesized FVIII-GFP in Weibel-Palade bodies, provide an attractive cellular on-demand delivery device for gene therapy of hemophilia A.


Subject(s)
Endothelial Cells/metabolism , Factor VIII/metabolism , Neovascularization, Physiologic , Calcimycin/pharmacology , Cell Line , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/physiology , Enzyme-Linked Immunosorbent Assay , Epinephrine/pharmacology , Factor VIII/genetics , Flow Cytometry , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Ionophores/pharmacology , Lentivirus/genetics , Microscopy, Confocal , Microscopy, Fluorescence , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transfection , Weibel-Palade Bodies/drug effects , Weibel-Palade Bodies/metabolism , von Willebrand Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...