scholarly journals Characterization of Leu777Pro and Ile865Thr type IIA von Willebrand disease mutations

Blood ◽  
1994 ◽  
Vol 83 (6) ◽  
pp. 1551-1557 ◽  
Author(s):  
SE Lyons ◽  
KA Cooney ◽  
P Bockenstedt ◽  
D Ginsburg

Abstract Type IIA von Willebrand disease (vWD) is an autosomal dominant bleeding disorder characterized by a qualitative defect in von Willebrand factor (vWF). A number of missense mutations responsible for type IIA vWD have recently been identified. This report examines the type IIA vWD mutations Leu777-->Pro and Ile865-->Thr by expression of recombinant vWF containing mutant and wild-type (WT) sequences. Recombinant vWF containing the L777P mutation (vWFL777P) showed markedly impaired secretion compared with that for wild-type vWF (vWFWT) after DNA transfection into mammalian cells. Multimer analysis of secreted vWFL777P showed predominantly low molecular weight forms. In contrast, recombinant vWF containing the I865T mutation (vWFI865T) was processed in a pattern similar to vWFWT, with secretion of the full spectrum of vWF multimers. Thus, L777P and I865T are subclassified as type IIA group I and group II mutations, respectively. Analysis of platelet vWF from a patient heterozygous for the L777P mutation shows reduced large vWF multimers in a pattern similar to plasma, consistent with the intracellular transport defect predicted for a group I mutation. An increase in the proportion of high molecular weight multimers observed in type IIA vWD patient plasma, after renal transplantation from a normal donor, suggests that the kidney endothelium may be a major source of plasma vWF.

Blood ◽  
1994 ◽  
Vol 83 (6) ◽  
pp. 1551-1557 ◽  
Author(s):  
SE Lyons ◽  
KA Cooney ◽  
P Bockenstedt ◽  
D Ginsburg

Type IIA von Willebrand disease (vWD) is an autosomal dominant bleeding disorder characterized by a qualitative defect in von Willebrand factor (vWF). A number of missense mutations responsible for type IIA vWD have recently been identified. This report examines the type IIA vWD mutations Leu777-->Pro and Ile865-->Thr by expression of recombinant vWF containing mutant and wild-type (WT) sequences. Recombinant vWF containing the L777P mutation (vWFL777P) showed markedly impaired secretion compared with that for wild-type vWF (vWFWT) after DNA transfection into mammalian cells. Multimer analysis of secreted vWFL777P showed predominantly low molecular weight forms. In contrast, recombinant vWF containing the I865T mutation (vWFI865T) was processed in a pattern similar to vWFWT, with secretion of the full spectrum of vWF multimers. Thus, L777P and I865T are subclassified as type IIA group I and group II mutations, respectively. Analysis of platelet vWF from a patient heterozygous for the L777P mutation shows reduced large vWF multimers in a pattern similar to plasma, consistent with the intracellular transport defect predicted for a group I mutation. An increase in the proportion of high molecular weight multimers observed in type IIA vWD patient plasma, after renal transplantation from a normal donor, suggests that the kidney endothelium may be a major source of plasma vWF.


1993 ◽  
Vol 70 (06) ◽  
pp. 1058-1062 ◽  
Author(s):  
Aida Inbal ◽  
Nurit Kornbrot ◽  
Paul Harrison ◽  
Anna M Randi ◽  
J Evan Sadler

SummaryType IIB von Willebrand disease (vWD) is characterized by a selective loss of high molecular weight von Willebrand factor (vWF) multimers in plasma due to their abnormally enhanced reactivity with platelets. Several missense mutations in the platelet glycoprotein lb (GPIb) binding domain of vWF were recently characterized that cause type IIB vWD. The effect of type IIB mutation Arg(545)Cys on vWF binding to platelet GPIb was studied using recombinant wild type (rvWFWT) and mutant rvWFR545C expressed in COS-7 cells. In the absence of ristocetin, 50% of rvWFR545C bound spontaneously to platelet GPIb and the binding increased to 70% in the presence of 0.2 mg/ml ristocetin; rvWFWT did not bind significantly under either condition. Botrocetin-induced binding of rvWFR545C was only slightly increased compared to rvWFWT. These data demonstrate that the Arg(545)Cys mutation increases the affinity of vWF for GPIb, resulting in the characteristic gain-of-function type IIB vWD phenotype.


1998 ◽  
Vol 79 (01) ◽  
pp. 211-216 ◽  
Author(s):  
Lysiane Hilbert ◽  
Claudine Mazurier ◽  
Christophe de Romeuf

SummaryType 2B of von Willebrand disease (vWD) refers to qualitative variants with increased affinity of von Willebrand factor (vWF) for platelet glycoprotein Ib (GPIb). All the mutations responsible for type 2B vWD have been located in the A1 domain of vWF. In this study, various recombinant von Willebrand factors (rvWF) reproducing four type 2B vWD missense mutations were compared to wild-type rvWF (WT-rvWF) for their spontaneous binding to platelets and their capacity to induce platelet activation and aggregation. Our data show that the multimeric pattern of each mutated rvWF is similar to that of WT-rvWF but the extent of spontaneous binding and the capacity to induce platelet activation and aggregation are more important for the R543Q and V553M mutations than for the L697V and A698V mutations. Both the binding of mutated rvWFs to platelets and platelet aggregation induced by type 2B rvWFs are inhibited by monoclonal anti-GPIb and anti-vWF antibodies, inhibitors of vWF binding to platelets in the presence of ristocetin, as well as by aurin tricarboxylic acid. On the other hand, EDTA and a monoclonal antibody directed against GPIIb/IIIa only inhibit platelet aggregation. Furthermore, the incubation of type 2B rvWFs with platelets, under stirring conditions, results in the decrease in high molecular weight vWF multimers in solution, the extent of which appears correlated with that of plasma vWF from type 2B vWD patients harboring the corresponding missense mutation. This study supports that the binding of different mutated type 2B vWFs onto platelet GPIb induces various degrees of platelet activation and aggregation and thus suggests that the phenotypic heterogeneity of type 2B vWD may be related to the nature and/or location of the causative point mutation.


Blood ◽  
2000 ◽  
Vol 96 (2) ◽  
pp. 560-568 ◽  
Author(s):  
Simon Allen ◽  
Adel M. Abuzenadah ◽  
Joanna Hinks ◽  
Joanna L. Blagg ◽  
Turkiz Gursel ◽  
...  

Abstract In this report we describe the molecular defect underlying partial and severe quantitative von Willebrand factor (VWF) deficiencies in 3 families previously diagnosed with types 1 and 3 Von Willebrand-disease. Analysis of the VWF gene in affected family members revealed a novel C to T transition at nucleotide 1067 of the VWF complemetary DNA (cDNA), predicting substitution of arginine by tryptophan at amino acid position 273 (R273W) of pre–pro-VWF. Two patients, homozygous for the R273W mutation, had a partial VWF deficiency (VWF:Ag levels of 0.06 IU/mL and 0.09 IU/mL) and lacked high-molecular weight VWF multimers in plasma. A third patient, also homozygous for the R273W mutation, had a severe VWF deficiency (VWF:Ag level of less than 0.01 IU/mL) and undetectable VWF multimers in plasma. Recombinant VWF having the R273W mutation was expressed in COS-7 cells. Pulse-chase experiments showed that secretion of rVWFR273W was severely impaired compared with wild-type rVWF. However, the mutation did not affect the ability of VWF to form dimers in the endoplasmic reticulum (ER). Multimer analysis showed that rVWFR273W failed to form high-molecular-weight multimers present in wild-type rVWF. We concluded that the R273W mutation is responsible for the quantitative VWF deficiencies and aberrant multimer patterns observed in the affected family members. To identify factors that may function in the intracellular retention of rVWFR273W, we investigated the interactions of VWF expressed in COS-7 cells with molecular chaperones of the ER. The R273W mutation did not affect the ability of VWF to bind to BiP, Grp94, ERp72, calnexin, and calreticulin in COS-7 cells.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2657-2657
Author(s):  
Andrea Artoni ◽  
Isabella Garagiola ◽  
Rossana Lombardi ◽  
Flora Peyvandi ◽  
Pier Mannuccio Mannucci

Abstract ADAMTS-13 cleaves high molecular weight von Willebrand factor (VWF) realeased by the endothelium in order to prevent massive intravascular platelets adhesion and aggregation as pathologically observed in thrombotic thrombocytopenic purpura. ADAMTS-13 is present at low levels in plasma. We therefore surmised that platelets are able to specifically bind the metalloprotease on their surface, hereby concentrating the enzyme where it is most required. With this as background, 96 wells polystyrene NUNC plates were coated either with albumin or fibrinogen or VWF or recombinant ADAMTS-13 (each at 10 μg/ml) and then blocked with 5% albumin overnight. Washed platelets (100,000/μl), incubated with divalent cations, were then let adhere to the wells for 1 hr at 37°C. After extensive washing adherent platelets were lysed, p-nitrophenilphosphate was added and the reaction was stopped with NaOH 2M. Detection was done by assessing optical density at 405 nm. Binding of washed platelets preincubated with 2mM CaCl2 and/or 2mM MgCl2 to wells covered with immobilized recombinant ADAMTS-13 was significantly higher than binding to wells coated with albumin (p<0.001), was at the same levels of the binding to wells covered with recombinant VWF and approximately half of binding to the wells coated with fibrinogen. When washed platelets were preincubated with EDTA 2mM binding to the wells coated with fibrinogen or recombinant ADAMTS-13 decreased at the same degree of the wells covered with albumin. Preincubation of platelets with antibodies against αIIbβ3 (7E3, 10 μg/ml) or GpIb (10 μg/ml) did not effect the levels of binding to recombinant ADAMTS-13 while the binding to fibrinogen and VWF was totally abolished. Activation of platelets, obtained by preincubating platelets with ADP (5 μM) or collagen (10 μg/ml), significantly increased their binding to recombinant ADAMTS-13 (p<0.001) as compared to the binding of non-activated platelets. Immunofluorescent studies were then performed to see whether or not ADAMTS-13 bound to the platelet plasma membrane using as primary antibody a murine anti-human ADAMTS-13 monoclonal antibody (13E2). A positive membrane fluorescent signal was detected using as a source of platelets either a normal donor or a patient with type III Von Willebrand disease, demonstrating that ADAMTS-13 is located on the platelet surface independently from VWF. In conclusion ADAMTS-13 binds to the surface of platelets, the binding is specific, activation and divalent cations dependent, inhibited by EDTA, and not mediated by VWF on the membrane of platelets; the binding site does not appear to be αIIbβ3 or GpIb. Hence it can be hypothesized that ADAMTS-13 on platelet membrane might cleave high molecular weight VWF.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2020-2020
Author(s):  
Cynthia M. Pruss ◽  
Carol A. Hegadorn ◽  
Andrea Labelle ◽  
Erin Burnett ◽  
Mia Golder ◽  
...  

Abstract Von Willebrand Factor (VWF) is a large multimeric glycoprotein that mediates platelet adhesion to the damaged blood vessel wall and subsequent platelet aggregation at the site of vascular injury. The size of VWF multimers is regulated by the metalloprotease, ADAMTS13. Alterations in VWF sequence can lead to an increase or decrease in ADAMTS13-mediated cleavage, resulting in either a loss or increase in high molecular weight VWF multimers, respectively. With the availability of a VWF knockout mouse, variant forms of VWF can be evaluated in vivo in terms of their contribution to hemostasis and thrombosis. In these studies, we have taken into account the significant differences in VWF-GPIb binding and ADAMTS13 cleavage efficiency seen between mice and humans and have also assumed that functionally important residues are likely to be conserved between species. With these considerations in mind, the protocols described in this report utilize mouse-exclusive reagents. Previous reports of correction of the VWF KO phenotype using hydrodynamic gene delivery have shown contradictory results for correction of the bleeding time and blood volume loss as well as a grossly abnormal multimer structure of the rescued VWF protein, due initially to an inadvertent C799R mutation and latterly to factors possibly related to the site of VWF synthesis. In this study, we compare wild type VWF clearance to a cleavage site knockout, Y1605A/M1606A, a type 2A Von Willebrand Disease (VWD) mutation, R1597W, and the common type 1 VWD mutation, Y1584C. The murine R1597W variant exhibited increased ADAMTS13-mediated cleavage (0.36- fold ADAMTS13 concentration), and the Y1605A/M1606A variant greatly decreased cleavage in vitro (>100-fold ADAMTS13 concentration). VWF KO mice, 7-10 weeks old, were injected with recombinant murine VWF (200U/kg) produced in HEK293 cells. VWF antigen levels (VWF:Ag), multimers, and complete blood counts (CBCs) were performed. Compared to the wild type infused protein (T1/2=33.2 minutes), the Y1605A/M1606A and R1597W mutant proteins show faster clearance (T1/2=17.7 minutes, p<0.001, and 27.5 minutes, p=0.025, respectively). Although, surprisingly, the Y1605A/M1606A variant showed a preferential loss of high molecular weight material compared to wild type recombinant protein, the R1597W type 2A mutant had a much more rapid loss of the high molecular multimers compared to the other proteins analyzed. No statistical differences were observed for platelet counts or other CBC parameters post protein infusion regardless of mutation compared to resting VWF KO mice. Hydrodynamic injections of a plasmid containing the ubiquitous synthetic CAG promoter and wild type VWF were performed on eight week old mice, with delivery of 100μg plasmid DNA in 10% body weight Ringer’s solution over 5-7 seconds. Maximum VWF:Ag levels of 10.67 U/ml were observed two days post infusion, with a significant proportion of observable high molecular weight VWF, indistinguishable from normal C57BL6 mouse plasma pool. Mouse FVIII:C levels follow a similar trend over the time course with maximum levels observed on day 3 at 206.7% activity. Mouse platelet counts were affected, with lower platelet counts rebounding to normal levels by day 7. No other changes in CBCs were observed. Intriguingly, none of the recombinant forms of VWF nor the hydrodynamically produced VWF protein show the typical triplet structure observed in normal mouse and human plasma, regardless of circulation time. The recombinant proteins migrate with the central band of the multimer triplet, while that of the hydrodynamic protein migrates with the lower triplet band, showing a lower molecular weight. These differences could be due to changes in protein structure or glycosylation from normally produced platelet and endothelial VWF. CAG-mVWF DNA Hydrodynamic Injection Results Days post infusion VWF:Ag (U/ml) FVIII:C (%) Platelets (103/μl) All data presented as value ± SD from 2-3 mice. Resting values are from at least 20 VWF KO mice. 1 4.82±0.18 62±16.7 440±101 2 10.67±1.06 128.5±53.6 547±30 3 9.92±0.53 206.7±29.4 558±39 7 1.03±0.47 78.3±3.3 699±53 10 0.51±0.53 30.4±16 502±189 14 0.13±0.08 26.8±0 470±276 Resting 0.0±0.0 (-) 569±167


Blood ◽  
2000 ◽  
Vol 96 (2) ◽  
pp. 560-568 ◽  
Author(s):  
Simon Allen ◽  
Adel M. Abuzenadah ◽  
Joanna Hinks ◽  
Joanna L. Blagg ◽  
Turkiz Gursel ◽  
...  

In this report we describe the molecular defect underlying partial and severe quantitative von Willebrand factor (VWF) deficiencies in 3 families previously diagnosed with types 1 and 3 Von Willebrand-disease. Analysis of the VWF gene in affected family members revealed a novel C to T transition at nucleotide 1067 of the VWF complemetary DNA (cDNA), predicting substitution of arginine by tryptophan at amino acid position 273 (R273W) of pre–pro-VWF. Two patients, homozygous for the R273W mutation, had a partial VWF deficiency (VWF:Ag levels of 0.06 IU/mL and 0.09 IU/mL) and lacked high-molecular weight VWF multimers in plasma. A third patient, also homozygous for the R273W mutation, had a severe VWF deficiency (VWF:Ag level of less than 0.01 IU/mL) and undetectable VWF multimers in plasma. Recombinant VWF having the R273W mutation was expressed in COS-7 cells. Pulse-chase experiments showed that secretion of rVWFR273W was severely impaired compared with wild-type rVWF. However, the mutation did not affect the ability of VWF to form dimers in the endoplasmic reticulum (ER). Multimer analysis showed that rVWFR273W failed to form high-molecular-weight multimers present in wild-type rVWF. We concluded that the R273W mutation is responsible for the quantitative VWF deficiencies and aberrant multimer patterns observed in the affected family members. To identify factors that may function in the intracellular retention of rVWFR273W, we investigated the interactions of VWF expressed in COS-7 cells with molecular chaperones of the ER. The R273W mutation did not affect the ability of VWF to bind to BiP, Grp94, ERp72, calnexin, and calreticulin in COS-7 cells.


Blood ◽  
1996 ◽  
Vol 87 (7) ◽  
pp. 2788-2794 ◽  
Author(s):  
T Englender ◽  
A Lattuada ◽  
PM Mannucci ◽  
JE Sadler ◽  
A Inbal

Type 2A von Willebrand disease (vWD), the most common qualitative form of vWD, is characterized by a relative decrease in circulating intermediate and high molecular weight (HMW) multimers. We studied the biosynthesis of recombinant von Willebrand factor (vWF) containing each of two type 2A vWD mutations previously reported by us, Arg834Gln and Val902Glu. The structure of recombinant Arg834Gln vWF within transfected COS-7 cells and the secretion of HMW multimers were similar to wild type vWF. The normal transport and secretion of Arg834Gln vWF, categorizes it as a group II type 2A mutation. In contrast, the Val90- 2Glu mutation resulted in intracellular proteolysis of vWF with the generation of a 176-kD fragment and retention of vWF between the endoplasmic reticulum and the Golgi complex. Moreover, the 176-kD fragment was also increased in plasma from patients with the Val902Glu mutation. Significantly impaired secretion and intracellular proteolysis of Val902Glu vWF categorizes a new sub-group of type 2A mutations. The intracellular proteolysis of vWF Val902Glu explains the lack of response to 1-deamino 8-D-arginine vasopressin (DDAVP) in patients who carry the mutation.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2708-2708
Author(s):  
Cynthia M. Pruss ◽  
Carol A. Hegadorn ◽  
Colleen R.P. Notley ◽  
Rouzbeh Chegeni ◽  
Aly S. Dhala ◽  
...  

Abstract Von Willebrand Factor (VWF) is a large multimeric glycoprotein that mediates platelet adhesion to the damaged blood vessel wall and subsequent platelet aggregation at the site of vascular injury. The size of VWF multimers in plasma is regulated by the specific VWF cleaving protease, ADAMTS13, that cleaves VWF at the Y1605-M1606 bond in the VWF A2 domain. The adhesive properties of VWF is directly related to the multimer size, with loss of high molecular weight VWF leading to the bleeding phenotype in Type 2A von Willebrand disease (VWD) and ultra high molecular weight VWF multimers observed in the prothrombotic state of thrombotic thrombocytopenic purpura (TTP). VWF mutations leading to multimer changes have not been examined in an animal model. Although the human and mouse VWF genes are highly conserved, mutations in the two genes have not been compared experimentally. To that end, we have made mutations in the mouse VWF (mVWF) cDNA to compare the ADAMTS13 cleavage patterns seen with human VWF mutations in two in vitro assays. Recombinant multimerized full-length mVWF was digested with different concentrations of recombinant mouse ADAMTS13 (mADAMTS13) and 1.5M urea, and analyzed via multimer migration distance. The GST and histidine-tagged G1554-T1668 A2 domain region of mVWF (mVWF115) was assayed via ELISA as follows: the mVWF115 was bound to anti-GST coated plates, digested with mADAMTS13, and intact mVWF115 detected via HRP-labeled anti-histidine tag antibody. We examined R1597W (VWD Type 2A), R1306W, (VWD Type 2B), Y1584C (VWD Type 1), as well as two previously described changes that drastically lower VWF cleavage, Y1605A/M1606A, and D1614A/E1615A/K1617A. Y1584C showed a statistically insignificant 41% decrease in cleavage in the full-length assay, but a 20% increase in the mVWF115 assay. Conversely, R1597W had a 64% decrease in the full-length assay, but a 28% increase in the mVWF115 assay. Y1605A/M1606A showed a dramatic loss of cleavage, with no observable loss of high molecular weight multimers at 32 units/ml of mADAMTS13, and a 126-fold higher mADAMTS13 concentration than wild type in the mVWF115 assay. This contrasted with the 8-fold increase observed with D1614A/E1615A/K1617A. The R1306W A1 domain mutation showed a significant increase in cleavage, with 63% less ADAMTS13 necessary in the full-length assay. The triple mutant R1306W/Y1605A/M1606A did not cleave at the highest concentration, 32 U/ml, similar to that of Y1605A/M1606A, showing that the R1306W mutation was not able to increase cleavage of the Y1605A/M1606A change. These results show a strong correlation between the mouse mutations described here and previously reported human VWF mutations in their susceptibility to ADAMTS13 cleavage. These results provide a rationale for the development of mouse models of type 1, 2A, and 2B VWD, as well as mVWF mutations that might lead to a prothrombotic state similar to TTP in humans. mADAMTS13 Concentration for 50% mVWF Cleavage ADAMTS13 Concentration (Units/ml) Relative mADAMTS13 Concentration (to Wild Type) mVWF Multimer (n=1) mVWF115 (n=4) mVWF Multimer (n=1) mVWF115 (n=4) *: P value <0.05, **: P Value < 0.001 in 2 tailed T test. Wild Type 0.301 0.197 1 1 Y1584C 0.423 0.157* 1.41 0.80* R1597W 0.109** 0.253* 0.36** 1.28* Y1605A/M1606A >32** 24.8** >100** 126** D1614A/E1615A/K1617A (−) 1.58** (−) 8.0** R1306W 0.112** (−) 0.37** (−) R1306W/Y1605A/M1606A >32** (−) >100** (−)


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 26-26
Author(s):  
Cynthia M. Pruss ◽  
Kate Sponagle ◽  
Kimberly Laverty ◽  
Carol A. Hegadorn ◽  
Yvette Chirinian ◽  
...  

Abstract Abstract 26 Introduction: Type 1 von Willebrand disease (VWD) is caused by mutations that result in moderate decreases in VWF (5-50% of normal levels) and a mild bleeding phenotype. The VWF missense mutation Y1584C is associated with mildly decreased VWF:Ag levels, increased ADAMTS13 cleavage, as well as a possible increase in clearance. The Vicenza mutation, R1205H, exhibits a more severe phenotype (VWF:Ag ∼10%), as well as accelerated clearance. However, extensive controlled in vitro and in vivo investigation of these mutations has yet to be described. In this study, we examine both Y1584C and R1205H in comparison to wild type VWF using in vitro and in vivo strategies employing both human and mouse VWF. Methods: Recombinant murine and human VWF and ADAMTS13 were produced via transient transfection in HEK293T cells in serum free OPTIMEM for 72 hours. Full length ADAMTS13 digests were performed in a Tris-Urea system and analyzed via multimer pattern. VWF115 digests were performed in an ELISA based assay. Hydrodynamic injections were performed using 100 μg wild type (WT) or mutant ET-mVWF plasmid DNA in Ringer's solution in 7-9 week old C57Bl6 VWF knockout mice. Mice were sampled at days 2, 5, 8, and then weekly. Mouse plasma was analyzed for CBCs, VWF:Ag, and VWF multimer structure. Results: In the HEK293T transient transfection system, secreted mutant protein was similar to that of wild type recombinant protein, with high molecular weight material present. ADAMTS13 digestion of full length recombinant Y1584C versus wild type showed no statistical difference: 50% cleavage for hVWF WT 1.78U hADAMTS13, hVWF Y1584C 1.67 U, P=0.58; mVWF WT 0.32 U mADAMTS13; Y1584C 0.42 U, P=0.11. In contrast, the Y1584C substitution in the hVWF115 construct required 40% less hADAMTS13 to effect equivalent cleavage (WT 0.087±0.014, 4; Y1584C 0.052±0.005, 8, mean U/ml ADAMTS13±SEM, N, P=0.013). Mouse ADAMTS13 cleavage of mVWF115 was also increased 20% for Y1584C (WT: 0.20±0.06, 4; Y1584C: 0.16±0.01, 4, P=0.014). Hydrodynamic injection caused no adverse events in any animals. CBC values were not statistically significantly different between wild type and mutants. Initial high VWF:Ag values were similar for wild type VWF (25.6±2.9, 15, mean U/ml±SEM, n) and Y1584C (27.2±5.0, 11), but R1205H levels were 36% lower (16.3±2.1, 10). At 14 days, WT VWF:Ag was 5.33±1.13, 15, with R1205H (1.73±0.44, 12) and Y1584C (1.84±0.59, 11) VWF:Ag levels being 68% and 65% lower, respectively. R1205H continued to remain approximately 40% of WT values for the next three weeks, while Y1584C continued to decrease, dropping to 15% on day 21 and 8% on day 28, compared to the wild type values at these time points. The R1205H mutation showed no significant difference in multimer structure defined by observed number of bands (days 2 to 42, mean difference -0.49, P>0.05) to wild type VWF. In contrast, Y1584C had a significant decrease in band number (3.38, P<0.001). Conclusions: This study demonstrates that these two type 1 VWD mutations have a strong observable effect in the VWF knockout mouse model. R1205H exhibits a large decrease in VWF:Ag levels at all measured time points, but no alteration in multimer structure. Y1584C, in contrast, shows a loss of high molecular weight material, and an initially high VWF:Ag level that rapidly decays from day 14 onward, suggesting increased ADAMTS13 cleavage and increased clearance. In addition, in vitro ADAMTS13 testing shows that Y1584C responds differently in the two assay systems with only the VWF115 assay showing significant increases in ADAMTS13-mediated cleavage. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document