Abstract
Terminal sialylation determines the plasma half-life of von Willebrand factor (VWF). A role for macrophage galactose lectin (MGL) in regulating hyposialylated VWF clearance has recently been proposed. In this study, we showed that MGL influences physiological plasma VWF clearance. MGL inhibition was associated with a significantly extended mean residence time and 3-fold increase in endogenous plasma VWF antigen levels (P<0.05). Using a series of VWF truncations, we further demonstrated that the A1 domain of VWF is predominantly responsible for enabling the MGL interaction. Binding of both full-length and VWF-A1-A2-A3 to MGL was significantly enhanced in the presence of ristocetin (P<0.05), suggesting that the MGL-binding site in A1 is not fully accessible in globular VWF. Additional studies using different VWF glycoforms demonstrated that VWF O-linked glycans, clustered at either end of the A1 domain, play a key role in protecting VWF against MGLmediated clearance. Reduced sialylation has been associated with pathological, increased clearance of VWF in patients with von Willebrand disease. Herein, we demonstrate that specific loss of α2-3 linked sialylation from O-glycans results in markedly increased MGL-binding in vitro, and markedly enhanced MGL-mediated clearance of VWF in vivo. Our data further show that the asialoglycoprotein receptor (ASGPR) does not have a significant role in mediating the increased clearance of VWF following loss of O-sialylation. Conversely however, we observed that loss of N-linked sialylation from VWF drives enhanced circulatory clearance predominantly via the ASGPR. Collectively, our data support the hypothesis that in addition to regulating physiological VWF clearance, the MGL receptor works in tandem with ASGPR to modulate enhanced clearance of aberrantly sialylated VWF in the pathogenesis of von Willebrand disease.
Introduction
von Willebrand disease (VWD) is the commonest inherited human bleeding disorder and is caused by either quantitative or qualitative deficiency of plasma von Willebrand factor (VWF).1,2 Increased plasma clearance of VWF constitutes an important mechanism in the pathogenesis of VWD.3 The MCMDM-1VWD European study, US Zimmerman Program and Willebrand in the Netherlands (WIN) study have all found pathological, enhanced VWF clearance in approximately 45% of patients with type 1 VWD, leading to the proposal that patients with a shortened VWF half-life should be considered as a distinct type 1C (1-Clearance) subgroup.4-8 Interestingly, subsequent studies have highlighted that enhanced VWF clearance also contributes to pathogenesis in patients with low VWF, as well as type 2 and type 3 VWD.7,9-12
Given the importance of enhanced clearance in the pathogenesis of VWD, significant research has focused on defining the cellular and molecular clearance pathways involved. Potential roles for macrophages, liver sinusoidal endothelial cells and hepatocytes have been proposed.13-17 A number of specific clearance receptors have also been described.3 These include the low-density lipoprotein receptor-related protein-1 (LRP1), the scavenger receptor class A member I (SR-A1), sialic-acid-bindingimmunoglobulin- like-lectins 5 (Siglec 5) and the macrophage galactose-type lectin (MGL) which are all expressed on macrophages.18-21 On liver sinusoidal endothelial cells, receptors that may play a role in VWF clearance include stabilin-2 (STAB2), scavenger receptor class A member 5 (SCARA 5) and C-type lectin domain family 4 member M (CLEC4M).17,22,23 Finally, the asialoglycoprotein receptor (ASGPR), predominantly expressed on hepatocytes and macrophages, has also been implicated.24
More than 30 different VWF sequence variations have been reported in patients with increased VWF clearance. 3,25 The archetypal type 1C mutation is the VWD Vicenza variant which is characterized by an R1205H substitution in the D3 domain of VWF.15,26 VWF glycosylation also plays a critical role in determining the rate of clearance of the protein.27-30 For example, plasma VWF:antigen (VWF:Ag) levels are 20-30% lower in blood group O individuals compared to non-O subjects due to a significant reduction in plasma half-life.31,32 Enzymatic removal of terminal sialic acid residues from VWF also markedly enhances clearance.27,28 Moreover, genetic inactivation of the ST3Gal-IV sialyltransferase was associated with a significant reduction in plasma VWF half-life.33 These data are important from a clinical perspective because the majority of both the N- and O-linked glycans of VWF are normally capped by sialic acid residues.34-37 As glycoproteins age in plasma, there is a stepwise elimination of saccharides from the termini of complex glycan chains.38 Glycan remodeling begins with loss of capping sialic acid, catalyzed by plasma neuraminadases 1 (Neu1) and 3 (Neu3) respectively. This time-dependent desialylation is important in triggering clearance of senescent glycoproteins. 38 Significantly reduced VWF sialylation levels have also been observed in a number of pathological conditions including sepsis, pulmonary hypertension and liver cirrhosis.39-41 Importantly, several groups have reported reduced VWF sialylation in patients with type 1 VWD.10,33,40,42 Together, these data suggest that quantitative sialylation plays a critical role in regulating both physiological and pathological clearance of VWF in vivo.
Grewal et al. originally described a role for the ASGPR in regulating enhanced clearance of desialylated VWF (particularly in the context of sepsis).24 More recently, we identified MGL as another receptor involved in regulating hyposialylated VWF clearance.21 Critically, however, important questions remain unanswered regarding the roles played by VWF sialylation in regulating physiological and/or pathological clearance. These include: (i) the relative importance of N- versus O-linked sialylation in regulating VWF clearance; (ii) the relative contributions of the ASGPR and MGL clearance receptors; and (iii) the molecular mechanisms through which hyposialylated VWF interacts with its clearance receptors.
Methods
A detailed description of the materials and methods can be found in the associated Online Supplementary Material.
Isolation and purification of human plasma-derived von Willebrand factor
Plasma-derived VWF (pdVWF) was purified from the VWFcontaining concentrate Fandhi® (Grifols, Barcelona, Spain) as previously described.21 Platelet-VWF was purified from lysed platelets as described elsewhere.43 Eluate fractions were then assessed for VWF antigen, multimer distribution, and purity.
Glycosidase digestion and quantitative analysis of glycan expression
To generate VWF glycoforms, pdVWF was treated with α2-3 neuraminidase, α2-3,6,8,9 neuraminidase, β1-3 galactosidase, peptide N-glycosidase F (PNGase F) and/or O-glycosidase under non-denaturing conditions overnight at 37°C.29,44 Following glycosidase digestion, changes in VWF glycans were assessed using specific lectin enzyme-linked immunosorbent assays (ELISA) as previously described.10
Expression and purification of recombinant von Willebrand factor variants
The expression vectors pcDNA-VWF encoding full length recombinant VWF, VWF-A1A2A3, VWF-A1, VWF-A2, VWF-A3, VWF-D’A3 or VWF-A3-CK fragments have previously been described.16 Additional VWF-A1 constructs containing either of the two O-linked glycan (OLG) clusters were also included; A1- OLG cluster 1 (T1248A, T1255A, T1256A, S1263A), and A1-OLG cluster 2 (T1468A, T1477A, S1486A, T1487A). All recombinant VWF variants were transiently expressed in HEK293T cells. Conditioned serum-free medium was harvested 72 h after transfection and concentrated via anion exchange chromatography as described before.16
In vitro von Willebrand factor binding studies
Solid phase plate-binding assays were used to evaluate VWF binding to MGL. Briefly, recombinant human MGL (Stratech, UK) was immobilized on a PolySorp™ 96-well plate (Nunc, Thermo Scientific™), the wells were blocked, and VWF was incubated at 37°C for 1 h. Bound VWF was detected using horseradish peroxidase (HRP)-conjugated polyclonal anti-VWF (Dako, Agilent Technologies), high-sensitivity streptavidin-HRP (ThermoScientific, UK) or anti-His-HRP antibody (Qiagen, UK) (see Online Supplementary Material for details).
von Willebrand factor clearance studies in MGL1-/-, VWF-/- and VWF-/-/Asgr1-/- mice
All clearance experiments were performed on mice 6 to 8 weeks old. All animal studies were approved by the Health Product Regulatory Authority, Ireland and an internal ethics committee. VWF-/- and Asgr1-/- mice, both on a C57BL/6J background, were obtained from the Jackson Laboratory (Sacremento, CA, USA) and crossbred to obtain a dual VWF-/-/Asgr1-/- knockout model as previously described.21 MGL specific clearance studies were also performed after inhibition of murine MGL1/2 using a commercial polyclonal goat anti-mouse MGL1/2 antibody (2 mg/kg) (R&D Systems, UK) as previously described.21 For endogenous clearance studies, murine VWF was labeled with N-hydroxysuccinimide-biotin (10 mg/kg; Thermo-Scientific), residual biotinylated murine VWF was quantified using a modified VWF ELISA. All clearance data were fitted to monoexponential equations, based on analysis of the Akaike information criterion. The slope and intercept of the equation of the line were used to calculate pharmacokinetic parameters including mean residence time (MRT) and half-life (t1/2).
Data presentation and statistical analysis
Experimental data were analyzed with GraphPad Prism version 8.0 (GraphPad Software, San Diego, CA, USA). Data were expressed as mean values ± standard error of the mean. Data were analyzed with the Student unpaired two-tailed t-test and P-values <0.05 were considered to be statistically significant.
Results
Physiological importance of MGL in regulating von Willebrand factor clearance in vivo
Mice have two distinct MGL homologs - murine MGL1 (mMGL1) and murine MGL2 (mMGL2).45 To gain insight into the biological importance of MGL, we first investigated murine MGL1 and MGL2 binding to VWF in vitro. Similar to human MGL, dose-dependent binding of both mMGL1 and mMGL2 to VWF was observed (Figure 1A). VWF binding to MGL2 was significantly greater than to mMGL1 (P<0.001). Murine plasma VWF:Ag levels are significantly elevated (~ 1.5 fold) in MGL1-/- mice. Since both mMGL1 and mMGL2 bind VWF, we hypothesized that knocking down mMGL1 alone may underestimate the biological importance of MGL-mediated clearance. Dual mMGL1-/-/mMGL2-/- mice are not commercially available; thus, to address this hypothesis, in vivo clearance studies were repeated in mMGL1-/- mice in the presence or absence of dual anti-MGL1/2 inhibitory antibodies. Following treatment with anti-MGL2, murine VWF:Ag levels were significantly increased compared to those in mMGL1-/- controls (2.78±04 U/mL vs. 1.5±0.5 U/mL respectively; P<0.05) (Figure 1B). Thus, complete murine MGL inhibition was associated with an almost 3-fold increase in endogenous plasma VWF:Ag levels compared to the levels in wild-type (mMGL1+/+mMGL2+/+) controls. In the presence of combined mMGL1 and mMGL2 inhibition, endogenous VWF clearance was significantly attenuated compared to that in controls (P<0.05) (Figure 1C) and murine VWF MRT was increased 2.4-fold (Figure 1D). These data confirm that the observed increase in murine VWF levels associated with inhibition of MGL-mediated clearance is attributable to an increase in VWF half-life. Importantly the magnitude of this effect of MGL on plasma VWF levels is also greater than that previously reported in Asgr1-/- mice (VWF:Ag levels increased ~1.5 fold).24 Collectively, these data demonstrate that mMGL2 constitutes another novel macrophage clearance receptor for VWF in mice. More importantly, the findings further suggest that MGL has a greater effect than ASGPR in regulating physiological VWF clearance.
The A domains of von Willebrand factor play a critical role in regulating MGL binding
Previous studies have demonstrated that macrophages play a key role in VWF clearance, and have further shown that macrophage receptor-recognition site(s) are present within the VWF-A1A2A3 domains.13,16 To assess the specific role of VWF domains in modulating MGL binding a series of variants were expressed (Figure 2A) (Online Supplementary Table S1). Since MGL is expressed on macrophages, we investigated whether the A domains of VWF influence MGL binding. Dose-dependent binding of pdVWF to recombinant human MGL was observed (Figure 2B). In keeping with the fact that MGL is a C-type lectin, this binding was ablated in the presence of EDTA (Figure 2B). Conversely, VWF-MGL binding was significantly enhanced in the presence of ristocetin (1 mg/mL) (Figure 2B). No significant effect of VWF multimer distribution on MGL-binding was observed (Online Supplementary Figure S1). Binding studies confirmed dose-dependent binding of VWF-A1A2A3 to MGL, which was again ristocetin- and calcium-dependent (Figure 2C). Finally, the relative importance of the individual domains within A1A2A3 in determining MGL binding was assessed. Although significant binding of the VWF-A1 domain to MGL was seen, no binding for either VWF-A2 or VWF-A3 was observed (Figure 2D). Together, these data support the hypothesis that the A1 domain of VWF plays a critical role in determining VWF binding to the MGL surface receptor on macrophages.
O-linked glycans on von Willebrand factor modulate the MGL interaction
Each VWF monomer contains 13 N-linked and ten Olinked glycan structures (Figure 3A).34,35 Whereas N-glycans are distributed across the VWF monomer, eight of the ten O-glycans are clustered in two groups around the VWF-A1 domain.35,36 To study the importance of specific N- and Oglycans in regulating MGL binding, pdVWF was treated with PNGase F and/or O-glycosidase respectively. Following each digestion, residual VWF glycan expression was assessed using lectin-binding ELISA (Online Supplementary Figure S2A, B). Although treatment with PNGase F successfully removed N-linked glycans from fulllength pdVWF, it did not have a significant effect on MGL binding (Figure 3B). Conversely, digestion with O-glycosidase was associated with a marked reduction in pdVWF binding to MGL (P<0.001) (Figure 3B). The VWF-A1A2A3 truncation contains two N-linked glycans and eight Olinked glycans (Figure 3A). In keeping with full-length VWF, PNGase treatment of A1A2A3-VWF (Online Supplementary Figure S2B, C) had no impact on the MGL interaction, whereas removal of O-glycans markedly attenuated binding (P<0.01) (Figure 3C). Digestion of isolated A1 domain with O-glycosidase (Online Supplementary Figure S2E) was also associated with a significant reduction in MGL binding (P<0.001) (Figure 3D). Finally, to investigate the relative importance of the two OLG clusters at either side of the A1 domain, isolated A1-OLG cluster 1 and A1-OLG cluster 2 were expressed (Figure 2A). Although MGL-binding was observed for both of these VWF A1 domain truncations, significantly enhanced binding was observed for A1-OLG cluster 2 (Figure 3E). Cumulatively, these findings demonstrate that VWF OLG, particularly those clustered either side of the A1 domain, play a major role in regulating MGL interaction.
α2-3 sialylation on O-glycans protects von Willebrand factor against MGL-mediated clearance
Recent mass spectrometry studies have characterized the O-glycan structures expressed on human pdVWF and highlighted significant heterogeneity (Figure 4A).35-37 Critically, however, a consistent feature of these O-glycan chains is that they generally terminate with sialic acid, which may be present in either α2-3 or α2-6 linkage.35,36 In contrast, most N-linked sialic acids are α2-6 linked.34 To further investigate the role of VWF O-glycans in determining MGL-mediated clearance, pdVWF was digested with a series of exoglycosidases to generate specific VWF glycoforms (Figure 4A). Treatment with α2-3 neuraminidase to remove α2-3 linked sialylation from O-glycans (Online Supplementary Figure S2F) significantly enhanced pdVWF binding to MGL (P=0.017) (Figure 4B). Similarly, digestion with α2-3,6,8,9 neuraminidase (which removes α2-3 linked sialylation from O-glycans and α2-6 linked sialylation from both N- and O-glycans) (Online Supplementary Figure S2G) was also associated with significantly increased MGL binding (P=0.006). Despite the fact that an estimated 80% of total sialylation on VWF is α2-6 linked, α2-3,6,8,9 Neu-VWF binding to MGL was not different to that observed following α2-3 neuraminidase digestion alone (Figure 4B). Significantly enhanced binding was observed for PNG-VWF following additional removal of α2-3 linked sialylation and exposure of the O-linked T antigen structure (Figure 4C). Finally, PNG-VWF was sequentially treated with α2-3 neuraminidase and β1-3 galactosidase to remove both terminal sialic acid and subterminal galactose (Gal) residues from VWF O-glycan chains (Online Supplementary Figure S2H). This combined digestion ablated the enhanced binding observed following α2-3 neuraminidase digestion alone (Figure 4C). These data demonstrate that α2-3 linked sialylation on VWF Oglycans specifically protects VWF against MGL-mediated clearance. Loss of this capping sialic acid results in Gal residue exposure on VWF O-glycans, which then triggers clearance through the MGL receptor. In order to consider whether other VWF domains/glycans may contribute to MGL-interaction, we compared binding for N-terminal D’A3-VWF and C-terminal A3-CK-VWF fragments. In keeping with a key role for the A1 domain, significant binding of D’A3-VWF to MGL was observed (Figure 4D). Interestingly, however, some A3-CK-VWF binding was also seen, suggesting that O-glycans (T1679 and/or T2298) downstream of the A1 domain may also play a role.
Role of MGL and ASGPR in modulating pathological enhanced clearance of desialylated von Willebrand factor
Previous studies have demonstrated altered VWF sialylation in patients with VWD as well as in a number of other conditions.41 To investigate the role of MGL in mediating the enhanced clearance of pathologically desialylated VWF, pdVWF was treated ex vivo with α2-3 neuraminidase to remove α2-3 linked sialylation from O-glycans. In vivo clearance studies were then performed in VWF-/- mice in the presence or absence of combined mMGL1 and mMGL2 inhibition. Removal of α2-3 linked sialylation was associated with a marked reduction in VWF half-life compared to that of the wild-type control (Figure 5A). Importantly, however, this enhanced clearance was attenuated in the presence of MGL inhibition (Figure 5A). To assess the relative roles of MGL and ASGPR in modulating the pathological, enhanced clearance following removal of α2-3 sialylation, in vivo clearance studies were also performed in dual VWF-/-Asgr1-/- knockout mice in the presence or absence of combined mMGL1 and mMGL2 inhibition (Figure 5B). Critically, we observed that MGL inhibition was also able to block enhanced clearance of pdVWF after loss of α2-3 sialylation equally effectively in the presence or absence of ASGPR (Figure 5B).
Terminal sialylation on VWF O-glycans can be either α2-3 or α2-6 linked. In contrast, sialylation on VWF Nglycan chains is predominantly α2-6 linked (Figure 4A).34,35 Since sepsis-related neuraminidases may target both the N- and O-glycans of VWF, we further investigated the role of MGL in clearing VWF from which both the N- and O-sialylation had been removed following digestion with α2-3,6,8,9 neuraminidase. In vivo clearance studies in VWF-/- mice demonstrated that combined mMGL1 and mMGL2 inhibition was not able to significantly reduce the pathological, enhanced clearance observed following loss of N-linked sialylation (Figure 6A). Interestingly, however, in mice deficient for the ASGPR clearance receptor, mMGL1/2 inhibition was associated with attenuation of the enhanced clearance of α2-3,6,8,9 Neu-VWF (Figure 6B). Collectively, these findings further support the hypothesis that O-linked α2-3 sialylation on VWF plays a critical role in protecting against MGL-mediated clearance. Moreover, the data also suggest that loss of α2-6 sialylation (predominantly Nlinked) on VWF drives enhanced clearance in a predominantly MGL-independent manner, mediated through the ASGPR.
Increased VWF clearance plays a key role in the pathogenesis of both type 1 and type 2B VWD.3,11,46 Previous studies have implicated macrophages, and in particular the LRP1 and SR-A1 receptors, in regulating this enanced clearance.14,15,19,20 To examine whether MGL may also play a role, we investigated binding for a number of type 1C (VWF-R1205H, R1205C, R1205S, S2179F) and type 2B (VWF-V1316M and -R1450E) variants. No evidence of enhanced MGL binding was observed for VWF-V1316M or any of the type 1C variants (Online Supplementary Figure S3). Interestingly, significantly reduced MGL binding was seen for VWF-R1450E compared to wild-type recombinant VWF. We hypothesise that this change in binding is due to conformational effects within the A1 domain affecting O-linked glycosylation during posttranslational modification and/or accessibility of specific OLG for the MGL interaction.
Platelet-von Willebrand factor sialylation and MGL interaction
Platelet α-granules contain approximatey 20% of the total VWF present in platelet-rich plasma.47,48 Previous studies have demonstrated that platelet-derived (plt)- VWF has altered glycosylation compared to pdVWF.48 In particular, plt-VWF does not express ABO blood group determinants and is hypo-sialylated.49,50 Importantly, these glycosylation differences influence susceptibility to ADAMTS-13 cleavage.43 Using lectin-binding ELISA, we confirmed that the quantitative reduction in plt-VWF sialylation was predominantly attributable to a specific reduction in N-linked sialylation (Figure 7A, B). As a result of this decreased N-sialylation, terminal galactose expression was significantly increased on plt-VWF compared to pd-VWF (Figure 7C). Critically, despite the significant reduction in N-linked sialylation, we observed no increase in MGL binding for plt-VWF (Figure 7D). Moreover, in vivo clearance of plt-VWF in VWF-/- mice was similar to that of pd-VWF (Figure 7E). Cumulatively, these novel data further support our hypothesis that Olinked sialylation on VWF plays a key role in protecting VWF against MGL-mediated clearance.
Discussion
Recent studies have demonstrated that complex glycan structures, which account for 20% of total VWF monomeric mass, play a key role in regulating the halflife of VWF in vivo.3,25,51 In addition, a number of lectin receptors have been shown to bind VWF.3 Critically, however, the relative importance of these receptors in modulating physiological and pathological VWF clearance has not been defined. Moreover, the particular VWF glycan determinants involved in modulating interactions with specific lectin receptors remain unclear. In this study, using a series of in vivo and in vitro methodologies, we demonstrated that both murine homologs of the MGL receptor bind to VWF and contribute to the physiological clearance of endogenous murine VWF. Consequently, combined inhibition of both mMGL1 and mMGL2 resulted in a 3-fold increase in murine plasma VWF levels which was attributable to a significant decrease in clearance rate. Importantly, the magnitude of the increased in vivo VWF levels associated with combined MGL inhibition was greater than that reported following inhibition of other VWF clearance receptors in mice (~2.5 fold vs. ~1.5 fold), suggesting that MGL plays an important role in regulating physiological clearance of VWF.
To further investigate how MGL interacts with VWF, we first investigated the roles of specific VWF domains. Our data demonstrate that the A1A2A3 domains of VWF are predominantly responsible for modulating MGL binding. Furthermore, studies using isolated A domains showed that the A1 domain plays a critical role in regulating the MGL interaction. Interestingly, the binding of both full-length and A1A2A3-VWF to MGL was markedly enhanced in the presence of ristocetin, suggesting that the MGL-binding site in A1 may not be fully accessible in normal globular VWF. This concept is in keeping with findings of previous studies that showed significantly increased VWF binding to macrophages in the presence of ristocetin, botrecetin or shear stress.14,16 From a biological perspective, these data suggest that any VWF circulating in an ‘active’ GpIb binding conformation will be cleared rapidly by macrophage MGL, which may be important in minimizing thrombotic risk. Importantly, our data further show that C-terminal A3-CK-VWF also binds MGL. Although the binding was less than that observed with N-terminal D’A3-VWF, this observation suggests that additional MGL-recognition sites beyond the A1 domain may contribute to the MGL interaction.
Mass spectrometry studies have demonstrated significant and site-specific heterogeneity in the carbohydrate structures expressed on human pdVWF.34-37 Nevertheless, the majority of both the N- and O-linked glycans are capped with negatively-charged sialic acid residues. In this study, we demonstrated that specific loss of α2-3 linked sialylation from the O-linked glycans of VWF causes enhanced MGL binding in vitro, and causes markedly enhanced MGL-mediated clearance in vivo. In contrast, removal of α2-6 linked sialylation, which constitutes most of the total sialic acid expressed on human VWF and, in particular, the vast majority of the sialylation on N-glycans, has minimal effect on MGL binding and/or clearance. Our data further suggest that the two O-linked glycan clusters located either side of the A1 domain play a key role in regulating binding to MGL. Previous studies have demonstrated that these O-glycan clusters have significant effects on local VWF conformation. 52,53 Further studies will be required to determine the molecular mechanisms through which these specific Oglycans regulate MGL-mediated VWF binding and clearance. Nevertheless, our findings demonstrate that MGL contributes to physiological VWF clearance by binding to exposed Gal residues on O-linked carbohydrate structures. Importantly, glycoprotein aging in plasma is associated with progressive loss of capping sialic acid, and thus increased exposure of these sub-terminal Gal residues.38
There are previous reports of significantly increased binding of RCA-I lectin to plasma VWF in patients with VWD.10,33,40,42 This lectin binds preferentially to Gal or GalNAc sugars which are typically present as sub-terminal residues on the O- and N-glycans of pdVWF, but become exposed following loss of capping sialic acid. Increased RCA-I binding has also been correlated with enhanced VWF clearance in VWD patients.10,33,40 Our data suggest that the shortened half-life associated with increased Gal exposure (and hence RCA-I binding) in VWD patients is mediated in large part through enhanced MGL-mediated clearance. Importantly, van Schooten et al. previously reported significantly increased binding of peanut agglutinin (PNA) lectin to VWF in a cohort of VWD patients.40 This lectin preferentially binds to the T antigen structure which is exposed following loss of Olinked sialylation. The authors further showed that increased PNA-binding (T antigen exposure) was associated with a significant increase in the VWFpp/VWF:Ag ratio, consistent with enhanced VWF clearance.40 In keeping with these results, we have demonstrated that α2-3 linked sialylation on O-linked glycan structures plays a particular role in protecting VWF against MGL-mediated clearance. Consequently, our findings suggest that the enhanced clearance associated with T antigen exposure on VWF previously reported by van Schooten et al. is attributable to enhanced clearance via MGL.
Besides VWD, abnormal VWF glycosylation has also been reported in a number of other disease states.24,39-41 For example, reduced PNA-binding to VWF has been reported in patients with liver cirrhosis who have significantly elevated plasma VWF:Ag levels. The biological mechanisms underlying reduced T antigen exposure on VWF in patients with cirrhosis have not been defined. Nonetheless, our findings build upon these previous observations and in particular suggest that the altered Oglycosylation associated with cirrhosis will cause increased plasma VWF levels as a result of decreased MGL-mediated clearance. Conversely, a number of different pathogens including Streptococcus pneumoniae, Haemophilus influenzae and Pseudomonas aeruginosa express neuraminidase enzymes that can cause desialylation of host glycoproteins.24,54 VWF desialylation associated with pathological, enhanced clearance has been observed in mice infected with S. pneumoniae.24 Our data further suggest that increased MGL-mediated clearance will play a key role in mediating this pathogen-associated enhanced VWF clearance. Interestingly, two previous studies have demonstrated that complete loss of Olinked carbohydrate structures is associated with significantly increased VWF clearance in vivo.28,55 Given that Oglycans are known to influence protein conformation, the observation that complete removal triggers enhanced clearance is likely attributable to conformational changes in VWF.
In addition to MGL, other macrophage receptors that can also interact with VWF include LRP1, SR-A1, Siglec- 5, Gal-1 and Gal-3.3,56,57 Some of these receptors have also been shown to bind with enhanced affinity to hyposialylated VWF (ASGPR, Gal-1 and Gal-3). Additional studies will be necessary to fully elucidate the relative roles of these other macrophage receptors in regulating the physiological and/or pathological clearance of hyposialylated VWF. Although it remains unclear whether these receptors may function synergistically in regulating desialylated VWF clearance, recent studies have demonstrated that LRP1 can form heterologous functional complexes with other macrophage receptors including β2-integrins. Importantly, Deppermann et al recently demonstrated that MGL on hepatic Küpffer cells plays a significant role in the removal of desialylated platelets, and that MGL and ASGPR appear to function collaboratively in physiological platelet clearance.58
Footnotes
- Received October 21, 2020
- Accepted March 12, 2021
Correspondence
Disclosures
JSO’D has served on speakers’ bureaux for Baxter, Bayer, Novo Nordisk, Boehringer Ingelheim, Leo Pharma, Takeda and Octapharma. He has also served on advisory boards for Baxter, Bayer, Octapharma CSL Behring, Daiichi Sankyo, Boehringer Ingelheim, Takeda and Pfizer and has received research grant funding awards from Baxter, Bayer, Pfizer, Shire, Takeda and Novo Nordisk. JMO’S has received research grant funding from LEO Pharma and Grifols
Contributions
SEW, ABM, JF and AC performed experiments; SEW, JMO’S, ABM, DS, RG, JS, JF, MM, TAJM, AC, SH and JSO’D designed the research and analyzed the data. All authors were involved in writing and reviewing the paper.
Data-sharing statement
All original data and protocols can be made available to other investigators upon request.
References
- Lenting PJ, Christophe OD, Denis CV. von Willebrand factor biosynthesis, secretion, and clearance: connecting the far ends. Blood. 2015; 125(13):2019-2028. https://doi.org/10.1182/blood-2014-06-528406PubMedGoogle Scholar
- Leebeek FWG, Eikenboom JCJ. von Willebrand's disease. N Engl J Med. 2017; 376(7):701-702. https://doi.org/10.1056/NEJMc1616060Google Scholar
- O'Sullivan JM, Ward S, Lavin M, O'Donnell JS. von Willebrand factor clearance - biological mechanisms and clinical significance. Br J Haematol. 2018; 183(2):185-195. https://doi.org/10.1111/bjh.15565PubMedGoogle Scholar
- Castaman G, Lethagen S, Federici AB. Response to desmopressin is influenced by the genotype and phenotype in type 1 von Willebrand disease (VWD): results from the European Study MCMDM-1VWD. Blood. 2008; 111(7):3531-3539. https://doi.org/10.1182/blood-2007-08-109231PubMedGoogle Scholar
- Flood VH, Christopherson PA, Gill JC. Clinical and laboratory variability in a cohort of patients diagnosed with type 1 VWD in the United States. Blood. 2016; 127(20):2481-2488. https://doi.org/10.1182/blood-2015-10-673681PubMedPubMed CentralGoogle Scholar
- Eikenboom J, Federici AB, Dirven RJ. VWF propeptide and ratios between VWF, VWF propeptide, and FVIII in the characterization of type 1 von Willebrand disease. Blood. 2013; 121(12):2336-2339. https://doi.org/10.1182/blood-2012-09-455089PubMedPubMed CentralGoogle Scholar
- Sanders YV, Groeneveld D, Meijer K. von Willebrand factor propeptide and the phenotypic classification of von Willebrand disease. Blood. 2015; 125(19):3006-3013. https://doi.org/10.1182/blood-2014-09-603241PubMedGoogle Scholar
- Haberichter SL, Castaman G, Budde U. Identification of type 1 von Willebrand disease patients with reduced von Willebrand factor survival by assay of the VWF propeptide in the European study: molecular and clinical markers for the diagnosis and management of type 1 VWD (MCMDM- 1VWD). Blood. 2008; 111(10):4979-4985. https://doi.org/10.1182/blood-2007-09-110940PubMedPubMed CentralGoogle Scholar
- Lavin M, Aguila S, Schneppenheim S. Novel insights into the clinical phenotype and pathophysiology underlying low VWF levels. Blood. 2017; 130(21):2344-2353. https://doi.org/10.1182/blood-2017-05-786699PubMedPubMed CentralGoogle Scholar
- Aguila S, Lavin M, Dalton N. Increased galactose expression and enhanced clearance in patients with low von Willebrand factor. Blood. 2019; 133(14):1585-1596. https://doi.org/10.1182/blood-2018-09-874636PubMedGoogle Scholar
- Casari C, Du V, Wu YP. Accelerated uptake of VWF/platelet complexes in macrophages contributes to VWD type 2Bassociated thrombocytopenia. Blood. 2013; 122(16):2893-2902. https://doi.org/10.1182/blood-2013-03-493312PubMedGoogle Scholar
- O'Donnell JS. Low VWF: insights into pathogenesis, diagnosis, and clinical management. Blood Adv. 2020; 4(13):3191-3199. https://doi.org/10.1182/bloodadvances.2020002038PubMedPubMed CentralGoogle Scholar
- Lenting PJ, Westein E, Terraube V. An experimental model to study the in vivo survival of von Willebrand factor. Basic aspects and application to the R1205H mutation. J Biol Chem. 2004; 279(13):12102-12109. https://doi.org/10.1074/jbc.M310436200PubMedGoogle Scholar
- van Schooten CJ, Shahbazi S, Groot E. Macrophages contribute to the cellular uptake of von Willebrand factor and factor VIII in vivo. Blood. 2008; 112(5):1704-1712. https://doi.org/10.1182/blood-2008-01-133181PubMedGoogle Scholar
- Rawley O, O'Sullivan JM, Chion A. von Willebrand factor arginine 1205 substitution results in accelerated macrophagedependent clearance in vivo. J Thromb Haemost. 2015; 13(5):821-826. https://doi.org/10.1111/jth.12875PubMedGoogle Scholar
- Chion A, O'Sullivan JM, Drakeford C. N-linked glycans within the A2 domain of von Willebrand factor modulate macrophage-mediated clearance. Blood. 2016; 128(15):1959-1968. https://doi.org/10.1182/blood-2016-04-709436PubMedGoogle Scholar
- Swystun LL, Lai JD, Notley C. The endothelial cell receptor stabilin-2 regulates VWF-FVIII complex half-life and immunogenicity. J Clin Invest. 2018; 128(9):4057-4073. https://doi.org/10.1172/JCI96400PubMedPubMed CentralGoogle Scholar
- Pegon JN, Kurdi M, Casari C. Factor VIII and von Willebrand factor are ligands for the carbohydrate-receptor Siglec-5. Haematologica. 2012; 97(12):1855-1863. https://doi.org/10.3324/haematol.2012.063297PubMedPubMed CentralGoogle Scholar
- Rastegarlari G, Pegon JN, Casari C. Macrophage LRP1 contributes to the clearance of von Willebrand factor. Blood. 2012; 119(9):2126-2134. https://doi.org/10.1182/blood-2011-08-373605PubMedGoogle Scholar
- Wohner N, Muczynski V, Mohamadi A. Macrophage scavenger receptor SR-AI contributes to the clearance of von Willebrand factor. Haematologica. 2018; 103(4):728-737. https://doi.org/10.3324/haematol.2017.175216PubMedPubMed CentralGoogle Scholar
- Ward SE, O'Sullivan JM, Drakeford C. A novel role for the macrophage galactosetype lectin receptor in mediating von Willebrand factor clearance. Blood. 2018; 131(8):911-916. https://doi.org/10.1182/blood-2017-06-787853PubMedGoogle Scholar
- Rydz N, Swystun LL, Notley C. The Ctype lectin receptor CLEC4M binds, internalizes, and clears von Willebrand factor and contributes to the variation in plasma von Willebrand factor levels. Blood. 2013; 121(26):5228-5237. https://doi.org/10.1182/blood-2012-10-457507PubMedPubMed CentralGoogle Scholar
- Swystun LL, Ogiwara K, Lai JD. The scavenger receptor SCARA5 is an endocytic receptor for von Willebrand factor expressed by littoral cells in the human spleen. J Thromb Haemost. 2019; 17(8):1384-1396. https://doi.org/10.1111/jth.14521PubMedPubMed CentralGoogle Scholar
- Grewal PK, Uchiyama S, Ditto D. The Ashwell receptor mitigates the lethal coagulopathy of sepsis. Nat Med. 2008; 14(6):648-655. https://doi.org/10.1038/nm1760PubMedPubMed CentralGoogle Scholar
- Casari C, Lenting PJ, Wohner N, Christophe OD, Denis CV. Clearance of von Willebrand factor. J Thromb Haemost. 2013; 11(Suppl 1):202-211. https://doi.org/10.1111/jth.12226PubMedGoogle Scholar
- Casonato A, Pontara E, Sartorello F. Reduced von Willebrand factor survival in type Vicenza von Willebrand disease. Blood. 2002; 99(1):180-184. https://doi.org/10.1182/blood.V99.1.180PubMedGoogle Scholar
- Sodetz JM, Pizzo SV, McKee PA. Relationship of sialic acid to function and in vivo survival of human factor VIII/von Willebrand factor protein. J Biol Chem. 1977; 252(15):5538-5546. https://doi.org/10.1016/S0021-9258(19)63384-3Google Scholar
- Stoddart JH, Andersen J, Lynch DC. Clearance of normal and type 2A von Willebrand factor in the rat. Blood. 1996; 88(5):1692-1699. https://doi.org/10.1182/blood.V88.5.1692.1692Google Scholar
- O'Sullivan JM, Aguila S, McRae E. Nlinked glycan truncation causes enhanced clearance of plasma-derived von Willebrand factor. J Thromb Haemost. 2016; 14(12):2446-2457. https://doi.org/10.1111/jth.13537PubMedGoogle Scholar
- Preston RJ, Rawley O, Gleeson EM, O'Donnell JS. Elucidating the role of carbohydrate determinants in regulating hemostasis: insights and opportunities. Blood. 2013; 121(19):3801-3810. https://doi.org/10.1182/blood-2012-10-415000PubMedGoogle Scholar
- Gallinaro L, Cattini MG, Sztukowska M. A shorter von Willebrand factor survival in O blood group subjects explains how ABO determinants influence plasma von Willebrand factor. Blood. 2008; 111(7):3540-3545. https://doi.org/10.1182/blood-2007-11-122945PubMedGoogle Scholar
- Ward SE, O'Sullivan JM, O'Donnell JS. The relationship between ABO blood group, von Willebrand factor, and primary hemostasis. Blood. 2020; 136(25):2864-2874. https://doi.org/10.1182/blood.2020005843PubMedPubMed CentralGoogle Scholar
- Ellies LG, Ditto D, Levy GG. Sialyltransferase ST3Gal-IV operates as a dominant modifier of hemostasis by concealing asialoglycoprotein receptor ligands. Proc Natl Acad Sci U S A. 2002; 99(15):10042-10047. https://doi.org/10.1073/pnas.142005099PubMedPubMed CentralGoogle Scholar
- Canis K, McKinnon TA, Nowak A. Mapping the N-glycome of human von Willebrand factor. Biochem J. 2012; 447(2):217-228. https://doi.org/10.1042/BJ20120810PubMedGoogle Scholar
- Canis K, McKinnon TA, Nowak A. The plasma von Willebrand factor O-glycome comprises a surprising variety of structures including ABH antigens and disialosyl motifs. J Thromb Haemost. 2010; 8(1):137-145. https://doi.org/10.1111/j.1538-7836.2009.03665.xPubMedGoogle Scholar
- Solecka BA, Weise C, Laffan MA, Kannicht C. Site-specific analysis of von Willebrand factor O-glycosylation. J Thromb Haemost. 2016; 14(4):733-746. https://doi.org/10.1111/jth.13260PubMedGoogle Scholar
- Gashash EA, Aloor A, Li D. An Insight into glyco-microheterogeneity of plasma von Willebrand factor by mass spectrometry. J Proteome Res. 2017; 16(9):3348-3362. https://doi.org/10.1021/acs.jproteome.7b00359PubMedPubMed CentralGoogle Scholar
- Yang WH, Aziz PV, Heithoff DM, Mahan MJ, Smith JW, Marth JD. An intrinsic mechanism of secreted protein aging and turnover. Proc Natl Acad Sci U S A. 2015; 112(44):13657-13662. https://doi.org/10.1073/pnas.1515464112PubMedPubMed CentralGoogle Scholar
- Lopes AA, Ferraz de Souza B, Maeda NY. Decreased sialic acid content of plasma von Willebrand factor in precapillary pulmonary hypertension. Thromb Haemost. 2000; 83(5):683-687. https://doi.org/10.1055/s-0037-1613892Google Scholar
- van Schooten CJ, Denis CV, Lisman T. Variations in glycosylation of von Willebrand factor with O-linked sialylated T antigen are associated with its plasma levels. Blood. 2007; 109(6):2430-2437. https://doi.org/10.1182/blood-2006-06-032706PubMedGoogle Scholar
- Ward S, O'Sullivan JM, O'Donnell JS. von Willebrand factor sialylation-A critical regulator of biological function. J Thromb Haemost. 2019; 17(7):1018-1029. https://doi.org/10.1111/jth.14471PubMedGoogle Scholar
- Millar CM, Riddell AF, Brown SA. Survival of von Willebrand factor released following DDAVP in a type 1 von Willebrand disease cohort: influence of glycosylation, proteolysis and gene mutations. Thromb Haemost. 2008; 99(5):916-924. https://doi.org/10.1160/TH07-09-0565PubMedGoogle Scholar
- McGrath RT, van den Biggelaar M, Byrne B. Altered glycosylation of plateletderived von Willebrand factor confers resistance to ADAMTS13 proteolysis. Blood. 2013; 122(25):4107-4110. https://doi.org/10.1182/blood-2013-04-496851PubMedGoogle Scholar
- McGrath RT, McKinnon TA, Byrne B. Expression of terminal alpha2-6-linked sialic acid on von Willebrand factor specifically enhances proteolysis by ADAMTS13. Blood. 2010; 115(13):2666-2673. https://doi.org/10.1182/blood-2009-09-241547PubMedGoogle Scholar
- Tsuiji M, Fujimori M, Ohashi Y. Molecular cloning and characterization of a novel mouse macrophage C-type lectin, mMGL2, which has a distinct carbohydrate specificity from mMGL1. J Biol Chem. 2002; 277(32):28892-28901. https://doi.org/10.1074/jbc.M203774200PubMedGoogle Scholar
- Wohner N, Legendre P, Casari C, Christophe OD, Lenting PJ, Denis CV. Shear stress-independent binding of von Willebrand factortype 2B mutants p.R1306Q & p.V1316M to LRP1 explains their increased clearance. J Thromb Haemost. 2015; 13(5):815-820. https://doi.org/10.1111/jth.12885PubMedGoogle Scholar
- Mannucci PM. Platelet von Willebrand factor in inherited and acquired bleeding disorders. Proc Natl Acad Sci U S A. 1995; 92(7):2428-2432. https://doi.org/10.1073/pnas.92.7.2428PubMedPubMed CentralGoogle Scholar
- McGrath RT, McRae E, Smith OP, O'Donnell JS. Platelet von Willebrand factor-- structure, function and biological importance. Br J Haematol. 2010; 148(6):834-843. https://doi.org/10.1111/j.1365-2141.2009.08052.xPubMedGoogle Scholar
- Williams SB, McKeown LP, Krutzsch H, Hansmann K, Gralnick HR. Purification and characterization of human platelet von Willebrand factor. Br J Haematol. 1994; 88(3):582-591. https://doi.org/10.1111/j.1365-2141.1994.tb05077.xPubMedGoogle Scholar
- Brown SA, Collins PW, Bowen DJ. Heterogeneous detection of A-antigen on von Willebrand factor derived from platelets, endothelial cells and plasma. Thromb Haemost. 2002; 87(6):990-996. https://doi.org/10.1055/s-0037-1613123Google Scholar
- Lenting PJ, Pegon JN, Christophe OD, Denis CV. Factor VIII and von Willebrand factor-- too sweet for their own good. Haemophilia. 2010; 16(Suppl 5):194-199. https://doi.org/10.1111/j.1365-2516.2010.02320.xPubMedGoogle Scholar
- Tischer A, Machha VR, Moon-Tasson L, Benson LM, Auton M. Glycosylation sterically inhibits platelet adhesion to von Willebrand factor without altering intrinsic conformational dynamics. J Thromb Haemost. 2020; 18(1):79-90. https://doi.org/10.1111/jth.14628PubMedPubMed CentralGoogle Scholar
- Deng W, Wang Y, Druzak SA. A discontinuous autoinhibitory module masks the A1 domain of von Willebrand factor. J Thromb Haemost. 2017; 15(9):1867-1877. https://doi.org/10.1111/jth.13775PubMedPubMed CentralGoogle Scholar
- Soong G, Muir A, Gomez MI. Bacterial neuraminidase facilitates mucosal infection by participating in biofilm production. J Clin Invest. 2006; 116(8):2297-2305. https://doi.org/10.1172/JCI27920PubMedPubMed CentralGoogle Scholar
- Badirou I, Kurdi M, Legendre P. In vivo analysis of the role of O-glycosylations of von Willebrand factor. PLoS One. 2012; 7(5):e37508. https://doi.org/10.1371/journal.pone.0037508PubMedPubMed CentralGoogle Scholar
- Saint-Lu N, Oortwijn BD, Pegon JN. Identification of galectin-1 and galectin-3 as novel partners for von Willebrand factor. Arterioscler Thromb Vasc Biol. 2012; 32(4):894-901. https://doi.org/10.1161/ATVBAHA.111.240309PubMedGoogle Scholar
- O'Sullivan JM, Jenkins PV, Rawley O. Galectin-1 and galectin-3 constitute novelbinding partners for factor VIII. Arterioscler Thromb Vasc Biol. 2016; 36(5):855-863. https://doi.org/10.1161/ATVBAHA.115.306915PubMedGoogle Scholar
- Deppermann C, Kratofil RM, Peiseler M. Macrophage galactose lectin is critical for Kupffer cells to clear aged platelets. J Exp Med. 2020; 217(4):e20190723. https://doi.org/10.1084/jem.20190723PubMedPubMed CentralGoogle Scholar
Data Supplements
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.