Interaction of ADAMTS13 with the Endothelial Cell Surface.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3710-3710 ◽  
Author(s):  
Anthony Vomund ◽  
Elaine M. Majerus

Abstract ADAMTS13 proteolysis of von Willebrand Factor (VWF) generates smaller multimers that are less likely to promote blood clotting. Deficiency of ADAMTS13 leads to thrombotic thrombocytopenic purpura, a frequently fatal disease, characterized by microangiopathic hemolytic anemia and thrombocytopenia. ADAMTS13 has a characteristic domain structure that includes metalloprotease and disintegrin domains, a thrombospondin type 1 repeat (TSR), cysteine-rich and spacer domains, 7 additional TSRs, and 2 carboxyl-terminal CUB domains. The ADAMTS13 substrate, VWF, is synthesized in endothelial cells and forms large multimers within the cell. These large multimers are secreted and adhere to the endothelial cell surface where they can bind platelets flowing in blood leading to thrombosis. ADAMTS13 has been shown to cleave VWF on the surface of endothelial cells, but it is unclear if ADAMTS13 also interacts with the endothelial cell surface. We have used iodinated ADAMTS13, fluorescence-activated cell sorting (FACS), and biochemical analysis using flow conditions to demonstrate that ADAMTS13 does interact with the endothelial cell surface. Iodinated ADAMTS13 bound the endothelial cell surface at 4oC. This binding was specific since the binding was inhibited in the presence of 40-fold excess unlabeled ADAMTS13. Binding of ADAMTS13 to the cell surface was time-dependent with maximal binding occurring within two hours. The binding was also reversible; the half-time for dissociation was four hours. Binding was inhibited by heparin but not by dextran sulfate. The Kd of binding to endothelial cells was 75 nM (range 40–100 nM). FACS analysis also demonstrated binding of ADAMTS13 to endothelial cells. A fluorescein isothiocyanate labeled anti-epitope antibody bound to endothelial cells in the presence but not the absence of ADAMTS13. A polyclonal antibody to VWF inhibited binding of ADAMTS13 to VWF, but this antibody did not affect binding of ADAMTS13 to endothelial cells, suggesting that ADAMTS13 interacts with endothelial cells independently of VWF. Studies with C-terminal truncation constructs of ADAMTS13 indicated that the carboxyl-terminal TSRs are important for binding since constructs terminating with the metalloprotease domain, the first TSR, or the sixth TSR failed to compete with full-length ADAMTS13 for binding to endothelial cells, but constructs terminating with either the seventh or eighth TSR did compete for binding. Lastly, recombinant ADAMTS13 was found to be associated with endothelial cells in flow experiments. Endothelial cells were perfused with medium containing plasma concentrations of ADAMTS13 (1 μg/ml) at 10 dynes/cm2. After perfusion, the endothelial cells were washed and bound ADAMTS13 was identified from whole cell lysates through SDS-PAGE and immunoblotting with an anti-V5 epitope antibody. ADAMTS13 was found associated with endothelial cells after perfusion. Binding of ADAMTS13 to the endothelial cells prior to perfusion led to enhanced proteolysis of VWF as compared to addition of ADAMTS13 during perfusion only. This suggests that the interaction of ADAMTS13 with endothelial cells is important since it enhances the cleavage of VWF as compared to that of ADAMTS13 in solution.

Blood ◽  
1993 ◽  
Vol 81 (3) ◽  
pp. 683-689 ◽  
Author(s):  
SR Lentz ◽  
JE Sadler

Abstract Intracellular protein transport in endothelial cells is selectively inhibited by homocysteine, a thiol amino acid associated with both thrombosis and atherosclerosis. In a previous study, homocysteine decreased cell surface expression of the surface transmembrane glycoprotein thrombomodulin without decreasing secretion of another endothelial cell protein, plasminogen activator inhibitor-1. To define further the effects of homocysteine on protein transport, we examined the processing and secretion of the multimeric glycoprotein von Willebrand factor (vWF) in human umbilical vein endothelial cells. Incubation with 2 mmol/L homocysteine resulted in complete loss of vWF multimers and prevented asparagine-linked oligosaccharide maturation, propeptide cleavage, and secretion; these effects are consistent with impaired exit from the endoplasmic reticulum (ER). Dimerization was only partially inhibited, suggesting that homocysteine causes retention of provWF in the ER without preventing dimer formation. In pulse-chase incubations, intracellular provWF was degraded before exiting the ER in homocysteine-treated cells. Homocysteine also inhibited the processing and secretion of a carboxyl-terminal truncation mutant of human provWF expressed in rat insulinoma cells, indicating that retention in the endoplasmic reticulum can be mediated by regions of provWF apart from the carboxyl-terminal 20-Kd segment. These results suggest that retention of secretory proteins in the ER is regulated by redox mechanisms and imply that the intracellular transport of multiple endothelial cell proteins may be altered in patients with homocystinuria.


Blood ◽  
1993 ◽  
Vol 81 (3) ◽  
pp. 683-689 ◽  
Author(s):  
SR Lentz ◽  
JE Sadler

Intracellular protein transport in endothelial cells is selectively inhibited by homocysteine, a thiol amino acid associated with both thrombosis and atherosclerosis. In a previous study, homocysteine decreased cell surface expression of the surface transmembrane glycoprotein thrombomodulin without decreasing secretion of another endothelial cell protein, plasminogen activator inhibitor-1. To define further the effects of homocysteine on protein transport, we examined the processing and secretion of the multimeric glycoprotein von Willebrand factor (vWF) in human umbilical vein endothelial cells. Incubation with 2 mmol/L homocysteine resulted in complete loss of vWF multimers and prevented asparagine-linked oligosaccharide maturation, propeptide cleavage, and secretion; these effects are consistent with impaired exit from the endoplasmic reticulum (ER). Dimerization was only partially inhibited, suggesting that homocysteine causes retention of provWF in the ER without preventing dimer formation. In pulse-chase incubations, intracellular provWF was degraded before exiting the ER in homocysteine-treated cells. Homocysteine also inhibited the processing and secretion of a carboxyl-terminal truncation mutant of human provWF expressed in rat insulinoma cells, indicating that retention in the endoplasmic reticulum can be mediated by regions of provWF apart from the carboxyl-terminal 20-Kd segment. These results suggest that retention of secretory proteins in the ER is regulated by redox mechanisms and imply that the intracellular transport of multiple endothelial cell proteins may be altered in patients with homocystinuria.


1987 ◽  
Author(s):  
Joost A Koedam ◽  
Jan J Sixma ◽  
Bonno N Bouma ◽  
David M Stern ◽  
Peter P Nawroth

Factor Xa (FXa) formation on the endothelial cell surface involves a membrane protein which promotes assembly of the Factor IXa-VIII-X complex. Since Factor VIII (FVIII) can also interact with von Willebrand factor (VWF), which is both present in the plasma and expressed by endothelium, we examined the effect of VWF on FXa formation. When monolayers of conditioned endothelium were incubated with FIXa (2.8 nM), FVIII (0.1 unit/ml), and FX (65 nM), the rate of FXa formation could be decreased in a dose-dependent manner by addition of VWF. At 10 min of incubation, a VWF concentration of 5 µg/ml caused a 93% inhibition of FXa formation. Addition of a polyclonal antibody (F(ab')2) directed against VWF which blocks formation of the FVIII-VwF complex, increased endothelial cell-dependent Factor IXa-VIII-mediated activation of FX by 2− to 3-fold in the absence of exogenous VWF, indicating a role for endogenous VWF. Since no VWF was detectable using a sensitive radioimmunoassay in reaction mixture supernatants, endothelial cell-associated VWF was considered as a potential binding site for FVIII, thereby removing it from the reaction mixture. In addition, we found no effect of either exogenous VWF nor anti VWF-antibodies when FVIII was activated with thrombin before starting the incubation.Radioligand binding studies were carried out with 125I-FVIII and demonstrated binding to a limited number of sites on intact endothelial cell monolayers which could be partially blocked by anti-VWF F(ab')2. These results suggest that VWF may regulate FXa formation on the endothelial cell surface.


2018 ◽  
Vol 2 (18) ◽  
pp. 2347-2357 ◽  
Author(s):  
Thejaswi Kalagara ◽  
Tracy Moutsis ◽  
Yi Yang ◽  
Karin I. Pappelbaum ◽  
Anne Farken ◽  
...  

Abstract The dynamic change from a globular conformation to an elongated fiber determines the ability of von Willebrand factor (VWF) to trap platelets. Fiber formation is favored by the anchorage of VWF to the endothelial cell surface, and VWF-platelet aggregates on the endothelium contribute to inflammation, infection, and tumor progression. Although P-selectin and ανβ3-integrins may bind VWF, their precise role is unclear, and additional binding partners have been proposed. In the present study, we evaluated whether the endothelial glycocalyx anchors VWF fibers to the endothelium. Using microfluidic experiments, we showed that stabilization of the endothelial glycocalyx by chitosan oligosaccharides or overexpression of syndecan-1 (SDC-1) significantly supports the binding of VWF fibers to endothelial cells. Heparinase-mediated degradation or impaired synthesis of heparan sulfate (HS), a major component of the endothelial glycocalyx, reduces VWF fiber–dependent platelet recruitment. Molecular interaction studies using flow cytometry and live-cell fluorescence microscopy provided further evidence that VWF binds to HS linked to SDC-1. In a murine melanoma model, we found that protection of the endothelial glycocalyx through the silencing of heparanase increases the number of VWF fibers attached to the wall of tumor blood vessels. In conclusion, we identified HS chains as a relevant binding factor for VWF fibers at the endothelial cell surface in vitro and in vivo.


Blood ◽  
2009 ◽  
Vol 114 (2) ◽  
pp. 425-436 ◽  
Author(s):  
Hong Yang ◽  
Sean Lang ◽  
Zhimin Zhai ◽  
Ling Li ◽  
Walter H. A. Kahr ◽  
...  

Abstract Platelet P-selectin plays important roles in inflammation and contributes to thrombosis and hemostasis. Although it has been reported that von Willebrand factor (VWF) affects P-selectin expression on endothelial cells, little information is available regarding regulation of platelet P-selectin expression. Here, we first observed that P-selectin expression was significantly decreased on platelets of fibrinogen and VWF double-deficient mice. Subsequently, we identified this was due to fibrinogen deficiency. Impaired P-selectin expression on fibrinogen-deficient platelets was further confirmed in human hypofibrinogenemic patients. We demonstrated that this impairment is unlikely due to excessive P-selectin shedding, deficient fibrinogen-mediated cell surface P-selectin binding, or impaired platelet granule release, but rather is due to decreased platelet P-selectin content. Fibrinogen transfusion completely recovered this impairment in fibrinogen-deficient (Fg−/−) mice, and engagement of the C-terminus of the fibrinogen γ chain with β3 integrin was required for this process. Furthermore, Fg−/− platelets significantly increased P-selectin expression following transfusion into β3 integrin–deficient mice and when cultured with fibrinogen. These data suggest fibrinogen may play important roles in inflammation, thrombosis, and hemostasis via enhancement of platelet P-selectin expression. Since human fibrinogen levels vary significantly in normal and diseased populations, P-selectin as an activation marker on platelets should be used with caution.


1990 ◽  
Vol 96 (2) ◽  
pp. 257-262
Author(s):  
C.J. Jackson ◽  
P.K. Garbett ◽  
B. Nissen ◽  
L. Schrieber

A major problem encountered when isolating human microvascular endothelium is the presence of contaminating cells such as fibroblasts that rapidly over-grow the endothelial cells. We describe here a simple, rapid technique for purifying endothelial cells derived from the microvasculature of neonatal foreskin and osteoarthritic and rheumatoid arthritic synovium. This technique is based on the selective binding of the lectin Ulex europaeus I (UEA I) to the endothelial cell surface via fucose residues. Initially UEA I was covalently bound to tosyl-activated super-paramagnetic polystyrene beads (Dynabeads) by incubation for 24 h at room temperature. Cells were isolated by extracting microvascular segments from enzyme-treated (trypsin and Pronase) cubes of tissue. The mixed population of cells obtained were purified by incubating them at 4 degrees C for 10 min with the UEA I-coated Dynabeads. Endothelium bound to the beads whilst contaminating cells were removed by five washes with HBSS using a magnetic particle concentrator. The endothelial cells thus obtained grew to confluence as a cobblestone-like monolayer and expressed von Willebrand factor antigen. The cells were released from the Dynabeads by the competitive binding of fucose (10 min at 4 degrees C). This new method is simple and reproducible and allows pure human microvascular endothelial cells to be cultured within 2 h of obtaining a specimen.


1987 ◽  
Author(s):  
J H Reinders ◽  
C L Verweii ◽  
J A V Mourlk ◽  
Ph G de Groot

Endothelial cells, cultured from human umbilical veins, synthesize von Willebrand Factor (vWF), that is stored by the cells in Weibel-Palade bodies, secreted into the medium and incorporated into the extracellular matrix underneath the cells. We have studied the influence of perturbation by phorbol esters and thrombin on the cellular distribution of vWF. Short-term (< 1 hour) treatment of endothelial cells with phorbol ester PMA or thrombin resulted in the release of cellular stored vWF. Long-term treatment with perturbants evoked a distinct change in the endothelial cell distribution of vWF, evident 24 to 48 hours after exposure. While the contents of the vWF storage vesicles were gradually restored within 48 hours, enhanced amounts of vWF were secreted into the medium. However, PMA did not increase the endothelial cell contents of mRNA encoding for vWF. The number as well as the size of vWF storage granules in the cells increased after exposure to perturbants. The perturbed cells responded to stimuli in releasing stored vWF, the amounts secreted were even greater than those in control cells. The extracellular matrix lost its vWF contents as the result of PMA or thrombin treatment, by blocking deposition of vWF in the matrix, not by enhancing degradation of matrix vWF. In perfusion experiments, the adhesion of washed platelets onto the isolated matrix of perturbed cells was considerable less than that in controls. Addition of vWF to the perfusate overcame this impairment. Thus, perturbation of endothelial cells changes the cellular distribution of vWF.Supported in part by ZWO grants 13-30-31 and 13-90-91 and Netherlands Heart Foundation grant 28.004.


2016 ◽  
Vol 6 (1) ◽  
Author(s):  
Francesco Ferraro ◽  
Mafalda Lopes da Silva ◽  
William Grimes ◽  
Hwee Kuan Lee ◽  
Robin Ketteler ◽  
...  

Abstract Changes in the size of cellular organelles are often linked to modifications in their function. Endothelial cells store von Willebrand Factor (vWF), a glycoprotein essential to haemostasis in Weibel-Palade bodies (WPBs), cigar-shaped secretory granules that are generated in a wide range of sizes. We recently showed that forcing changes in the size of WPBs modifies the activity of this cargo. We now find that endothelial cells treated with statins produce shorter WPBs and that the vWF they release at exocytosis displays a reduced capability to recruit platelets to the endothelial cell surface. Investigating other functional consequences of size changes of WPBs, we also report that the endothelial surface-associated vWF formed at exocytosis recruits soluble plasma vWF and that this process is reduced by treatments that shorten WPBs, statins included. These results indicate that the post-exocytic adhesive activity of vWF towards platelets and plasma vWF at the endothelial surface reflects the size of their storage organelle. Our findings therefore show that changes in WPB size, by influencing the adhesive activity of its vWF cargo, may represent a novel mode of regulation of platelet aggregation at the vascular wall.


1987 ◽  
Author(s):  
Freek van Iwaarden ◽  
G Philip ◽  
de Groot ◽  
Bonno N Bouma

The presence of High Molecular Weight kininogen (HMWK) was demonstrated in cultured human endothelial cells (EC) by immunofluorescence techniques. Using an enzyme linked immunosorbent assay a concentration of 58 ng HMWK/10 cells was determined. Immunoprecipitation studies performed with lysed metabolically labelled endothelial cells and mono-specific antisera directed against HMWK suggested that HMWK is not synthesized by the endothelial cells. Endothelial cells cultured in the presence of HMWK-depleted serum did not contain HMWK. This, suggests that endothelial cells can internalize HMWK. Using 125I-HMWK it was demonstrated that cultured endothelial cells bind HMWK in a time-dependent, specific and saturable.way. The cells were found to internalize 125I-HMWK, since I-HMWK was detected in solubilized endothelial cells after the cell bound 125I-HMWK had been eluted with dextran sulphate.The binding of I-HMWK required the presence of zinc ions. Optimal binding of 125I-HMWK was observed at 50 μM Zn++ . Calcium ions inhibited the Zn++ dependent binding of 125I-HMWK |25EC. In the presence of 3 mM CaCl2 the total binding of 125I-HMWK was significantly decreased, and a .concentration of 200 μM Zn++ was Required for the binding of 125I-HMWK to thecells. Higher,. Ca concentrations did not further decrease the binding of 125I-HMWK. Analysis of tl^e binding data by the ligand computer program indicated 3.2 x 10 binding sites per cell for HMWK with a Kd of 35 nM at 50 μM ZnCl2 and 1 mM CaCl2. Specify binding of HMWK did also occur at physiological plasma Zn++ concentrations. Half maximal binding was observed at HMWK concentrations of ± 105 nM at 10 μM ZnCl2 and 45 nM at 25 μM ZnCl2. The HMWK binding sites were saturatecT at HMWK concentrations of 130 nM with 1.6 x 10 molecules of HMWK bound per cell and at 80 nM with 2.8 x 10 molecules of HMWK bound per cell at 10 and 25 pM ZnCl2 respectively. These results suggest that at physiological zinc, calcium and HMWK concentrations the HMWK binding sites on the endothelial cell are saturated. The presence of HMWK on the endothelial cell surface may play a role in the initiation of the intrinsic coagulation pathway. M ZnCl2 and 45 nM at 25 μM ZnCl2. The HMWK binding sites were saturatecT at HMWK concentrations of 130 nM with 1.6 x 10 molecules of HMWK bound per cell and at 80 nM with 2.8 x 10 molecules of HMWK bound per cell at 10 and 25 μM ZnCl2 respectively. These results suggest that at physiological zinc, calcium and HMWK concentrations the HMWK binding sites on the endothelial cell are saturated. The presence of HMWK on the endothelial cell surface may play a role in the initiation of the intrinsic coagulation pathway. M ZnCl2 and 45 nM at 25 μM ZnCl2. The HMWK binding sites were saturatecT at HMWK concentrations of 130 nM with 1.6 x 10 molecules of HMWK bound per cell and at 80 nM with 2.8 x 10 molecules of HMWK bound per cell at 10 and 25 μM ZnCl2 respectively. These results suggest that at physiological zinc, calcium and HMWK concentrations the HMWK binding sites on the endothelial cell are saturated. The presence of HMWK on the endothelial cell surface may play a role in the initiation of the intrinsic coagulation pathway. M ZnCl2 and 45 nM at 25 μM ZnCl2. The HMWK binding sites were saturatecT at HMWK concentrations of 130 nM with 1.6 x 10 molecules of HMWK bound per cell and at 80 nM with 2.8 x 10 molecules of HMWK bound per cell at 10 and 25 μM ZnCl2 respectively. These results suggest that at physiological zinc, calcium and HMWK concentrations the HMWK binding sites on the endothelial cell are saturated. The presence of HMWK on the endothelial cell surface may play a role in the initiation of the intrinsic coagulation pathway.M ZnCl2 and 45 nM at 25 μM ZnCl2. The HMWK binding sites were saturatecT at HMWK concentrations of 130 nM with 1.6 x 16 molecules of HMWK bound per cell and at 80 nM with 2.8 x 106 molecules of HMWK bound per cell at 10 and 25 μM ZnCl2 respectively. These results suggest that at physiological zinc, calcium and HMWK concentrations the HMWK binding sites on the endothelial cell are saturated. The presence of HMWK on the endothelial cell surface may play a role in the initiation of the intrinsic coagulation pathway.


2020 ◽  
Vol 10 (1) ◽  
pp. 31 ◽  
Author(s):  
Smart Ikechukwu Mbagwu ◽  
Luis Filgueira

Cerebral microvascular endothelial cells (CMVECs) line the vascular system of the brain and are the chief cells in the formation and function of the blood brain barrier (BBB). These cells are heterogeneous along the cerebral vasculature and any dysfunctional state in these cells can result in a local loss of function of the BBB in any region of the brain. There is currently no report on the distribution and variation of the CMVECs in different brain regions in humans. This study investigated microcirculation in the adult human brain by the characterization of the expression pattern of brain endothelial cell markers in different brain regions. Five different brain regions consisting of the visual cortex, the hippocampus, the precentral gyrus, the postcentral gyrus, and the rhinal cortex obtained from three normal adult human brain specimens were studied and analyzed for the expression of the endothelial cell markers: cluster of differentiation 31 (CD31) and von-Willebrand-Factor (vWF) through immunohistochemistry. We observed differences in the expression pattern of CD31 and vWF between the gray matter and the white matter in the brain regions. Furthermore, there were also regional variations in the pattern of expression of the endothelial cell biomarkers. Thus, this suggests differences in the nature of vascularization in various regions of the human brain. These observations also suggest the existence of variation in structure and function of different brain regions, which could reflect in the pathophysiological outcomes in a diseased state.


Sign in / Sign up

Export Citation Format

Share Document