GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    Online Resource
    Online Resource
    American Society of Hematology ; 2016
    In:  Blood Vol. 128, No. 22 ( 2016-12-02), p. 1430-1430
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 1430-1430
    Abstract: Background: Splanchnic vein thrombosis (SVT) includes extrahepatic portal vein obstruction (EHPVO), Budd-Chiari syndrome (BCS) and mesenteric vein thrombosis. Myeloproliferative neoplasms (MPN) account for the majority of non-cirrhotic SVT and are diagnosed in 30% of patients with EHPVO and 40% with BCS. Recurrent thrombosis is a common and significant complication in patients with PVT and MPN. Causes of thrombosis in this patient group are unknown; it is likely multifactorial, with proposed risk factors including increased platelet mass and platelet hyper-reactivity. There is evidence that in general, patients with MPN have increased platelet reactivity when compared to healthy controls, but, there is no data looking specifically at patients with SVT. However patients with SVT clearly demonstrate a significantly more aggressive phenotype in terms of systemic thrombosis, with risk of recurrent thrombotic episodes. MASCOT is a multicenter observational study assessing morbidity and portal circulation in patients with SVT, in a subset of whom we have assessed platelet activity. Methods: Whole blood flow cytometry was used to assess platelet activity by surface expression of P-selectin (CD62P) an established marker of platelet activity. We measured CD62P at baseline in unstimulated platelets and following stimulation with increasing concentrations of thrombin receptor activator protein (TRAP), range 10-50μmol. The guidelines from the European consensus on platelet flow cytometry were used for this assay (Schmitz G et al 1998). Demographic and clinical data were collected. Results were analysed in GraphPad Prism using t-tests; all values displayed as mean (95% confidence interval). Results: We assessed platelet activity in 14 patients with SVT using healthy controls (n=6) for comparison. In our patient cohort 8/14 (57%) were male and 6/14 (42%) female. The average age of our patients was 49 years. 12/14 (85%) were positive for the JAK2 mutation and 2/14 (15%) had a positive calreticulin mutation (CALR+). 2/14 (14%) patients had myelofibrosis (MF), 6/14 (42%) polycythaemia vera (PV), 4/14 (28%) essential thrombocytosis (ET) and 2/14 (14%) were positive for the JAK2 mutation but did not show evidence of MPN in their bone marrow and had normal blood counts. 10/14 (71%) were on warfarin alone, 2/14 (14%) on warfarin and aspirin, 1/14 (7%) on rivaroxaban and 1/14 (7%) on aspirin alone. Platelet activity as measured by CD62P expression was increased in patients with SVT at baseline compared with controls 13.1% (5.5-20.7) vs 0.5% (0.2-0.7) (p=0.003). Platelet activation and reactivity was significantly greater in the patient group compared with controls at all concentrations of TRAP (figure 1). Control platelets were relatively unreactive to trap stimulation and a significant increase from baseline in CD62P at the highest concentration of TRAP used (50 μmol) 0.5% (0.2-0.7) vs 7.31% (1.17-13.4) (p=0.03), however this is a modest increase, the biological significance of which is uncertain. In the patient population, the platelets were hyper-reactive to TRAP induced up-regulation of CD62P. This was significant when comparing unstimulated platelets at the 25 and 50 μmol concentrations 13.1% (5.5-20.7) vs 22.6% (13.8-39.4) (p=0.0008) and 13.1% (5.5-20.7) vs 34.9% (22.6-47.1) (p 〈 0.0001) respectively. Although not significant at the lower concentration of 10 μmol TRAP there was a trend towards significance 13.1% (5.5-20.7) vs 22% (12.1-38.9) (p=0.07). Discussion: These results demonstrate that patients with SVT have both significantly increased baseline markers of platelet activation and increased reactivity to stimulation. As part of this longitudinal study, we propose to monitor changes in platelet activity in this patient cohort over time to assess whether or not changes in treatment modality influence platelet activity and whether or not changes in platelet activity can predict further thrombotic episodes; this data will be available in the coming year. Our data provides a platform for further investigation; in due course we will control this data using patients with MPN without thrombosis, as further patients within our study group are analysed. Disclosures Chen: Novartis: Other: Advisory Board. Sekhar:Novartis: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Society of Hematology ; 2006
    In:  Blood Vol. 108, No. 11 ( 2006-11-16), p. 1701-1701
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 1701-1701
    Abstract: Von Willebrand factor (VWF) is a large plasma glycoprotein that mediates platelet tethering at sites of vascular injury. This function is dependent upon its multimeric size, which is modulated in plasma through proteolysis by ADAMTS13. Terminal ABO blood group sugars presented on the N-linked glycans of VWF alter the susceptibility of VWF to cleavage by ADAMTS13. Two predicted N-linked glycan sites (N1515 & N1574) are in close proximity to the ADAMTS13 cleavage site (Y1605-M1606). However, is it not known whether these predicted sites are occupied, or indeed, present the ABO sugars. In this study, purified plasma VWF was digested with trypsin, and the resulting fragments separated by ion-exchange chromatography. A 55kDa fragment observed under reducing SDS-PAGE was identified as VWF residues 1493–1849 by mass spectrocosopy and N-terminal sequencing. This fragment encompassed the predicted N-linked glycan sites at N1515 & N1574. Incomplete PNGase F digestion produced three protein bands, indicating that both glycosylation sites were occupied. Furthermore, analysis with blood group specific lectins demonstrated presentation of the ABO sugars on these glycan chains. This data suggests that the N1515 and N1574 glycosylation sites and subsequent ABO(H) sugar modification may influence VWF proteolysis. To further elucidate the role of other VWF glycan structures on susceptibility to ADAMTS13 cleavage, purified VWF was treated with neuraminidase to remove terminal sialic acid residues, producing asialo VWF (As-VWF), and PNGase F to remove whole N-linked glycan chains. (Nless-VWF). Lectin analysis demonstrated removal of 〉 95% of terminal sialic residues and 〉 75% whole N-linked glycan chains. Both AsVWF and Nless-VWF bound to type III collagen with similar affinity to wild type VWF (KD 1.9nM, 1.6nM, 1.4nM respectively) and demonstrated similar multimeric composition. Interestingly, As-VWF had decreased susceptibility to ADAMTS13 cleavage, but bound ADAMTS13 with comparable affinity to wild type VWF (KD 11nM & 12nM respectively). Nless-VWF was cleaved faster by ADAMTS13 and bound ADAMTS13 with ~ 4-fold increased affinity (KD 3.4nM). This data demonstrates that the glycan moiety of VWF plays an important role in mediating the interaction with ADAMTS13.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 117, No. 3 ( 2011-01-20), p. 1071-1080
    Abstract: The regulation of blood vessel formation is of fundamental importance to many physiological processes, and angiogenesis is a major area for novel therapeutic approaches to diseases from ischemia to cancer. A poorly understood clinical manifestation of pathological angiogenesis is angiodysplasia, vascular malformations that cause severe gastrointestinal bleeding. Angiodysplasia can be associated with von Willebrand disease (VWD), the most common bleeding disorder in man. VWD is caused by a defect or deficiency in von Willebrand factor (VWF), a glycoprotein essential for normal hemostasis that is involved in inflammation. We hypothesized that VWF regulates angiogenesis. Inhibition of VWF expression by short interfering RNA (siRNA) in endothelial cells (ECs) caused increased in vitro angiogenesis and increased vascular endothelial growth factor (VEGF) receptor-2 (VEGFR-2)–dependent proliferation and migration, coupled to decreased integrin αvβ3 levels and increased angiopoietin (Ang)–2 release. ECs expanded from blood-derived endothelial progenitor cells of VWD patients confirmed these results. Finally, 2 different approaches, in situ and in vivo, showed increased vascularization in VWF-deficient mice. We therefore identify a new function of VWF in ECs, which confirms VWF as a protein with multiple vascular roles and defines a novel link between hemostasis and angiogenesis. These results may have important consequences for the management of VWD, with potential therapeutic implications for vascular diseases.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 114, No. 16 ( 2009-10-15), p. 3489-3496
    Abstract: Investigation of 3 families with bleeding symptoms demonstrated a defect in the collagen-binding activity of von Willebrand factor (VWF) in association with a normal VWF multimeric pattern. Genetic analysis showed affected persons to be heterozygous for mutations in the A3 domain of VWF: S1731T, W1745C, and S1783A. One person showed compound heterozygosity for W1745C and R760H. W1745C and S1783A have not been reported previously. The mutations were reproduced by site-directed mutagenesis and mutant VWF expressed in HEK293T cells. Collagen-binding activity measured by immunosorbent assay varied according to collagen type: W1745C and S1783A were associated with a pronounced binding defect to both type I and type III collagen, whereas the principal abnormality in S1731T patients was a reduction in binding to type I collagen only. The multimer pattern and distribution of mutant proteins were indistinguishable from wild-type recombinant VWF, confirming that the defect in collagen binding resulted from the loss of affinity at the binding site and not impairment of high-molecular-weight multimer formation. Our findings demonstrate that mutations causing an abnormality in the binding of VWF to collagen may contribute to clinically significant bleeding symptoms. We propose that isolated collagen-binding defects are classified as a distinct subtype of von Willebrand disease.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 111, No. 7 ( 2008-04-01), p. 3407-3414
    Abstract: We report a 3-generation pedigree with 5 individuals affected with a dominantly inherited macrothrombocytopenia. All 5 carry 2 nonsynonymous mutations resulting in a D723H mutation in the β3 integrin and a P53L mutation in glycoprotein (GP) Ibα. We show that GPIbα-L53 is phenotypically silent, being also present in 3 unaffected pedigree members and in 7 of 1639 healthy controls. The β3-H723 causes constitutive, albeit partial, activation of the αIIbβ3 complex by disruption of the highly conserved cytoplasmic salt bridge with arginine 995 in the αIIb integrin as evidenced by increased PAC-1 but not fibrinogen binding to the patients' resting platelets. This was confirmed in CHO αIIbβ3-H723 transfectants, which also exhibited increased PAC-1 binding, increased adhesion to von Willebrand factor (VWF) in static conditions and to fibrinogen under shear stress. Crucially, we show that in the presence of fibrinogen, αIIbβ3-H723, but not wild-type αIIbβ3, generates a signal that leads to the formation of proplatelet-like protrusions in transfected CHO cells. Abnormal proplatelet formation was confirmed in the propositus's CD34+ stem cell–derived megakaryocytes. We conclude that the constitutive activation of the αIIbβ3-H723 receptor causes abnormal proplatelet formation, leading to incorrect sizing of platelets and the thrombocytopenia observed in the pedigree.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 121, No. 14 ( 2013-04-04), p. 2773-2784
    Abstract: BOECs from VWD patients provide novel insight into the cellular mechanisms of the disease.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 133, No. 24 ( 2019-06-13), p. 2559-2569
    Abstract: During wound healing, the distribution, availability, and signaling of growth factors (GFs) are orchestrated by their binding to extracellular matrix components in the wound microenvironment. Extracellular matrix proteins have been shown to modulate angiogenesis and promote wound healing through GF binding. The hemostatic protein von Willebrand factor (VWF) released by endothelial cells (ECs) in plasma and in the subendothelial matrix has been shown to regulate angiogenesis; this function is relevant to patients in whom VWF deficiency or dysfunction is associated with vascular malformations. Here, we show that VWF deficiency in mice causes delayed wound healing accompanied by decreased angiogenesis and decreased amounts of angiogenic GFs in the wound. We show that in vitro VWF binds to several GFs, including vascular endothelial growth factor-A (VEGF-A) isoforms and platelet-derived growth factor-BB (PDGF-BB), mainly through the heparin-binding domain (HBD) within the VWF A1 domain. VWF also binds to VEGF-A and fibroblast growth factor-2 (FGF-2) in human plasma and colocalizes with VEGF-A in ECs. Incorporation of the VWF A1 HBD into fibrin matrices enables sequestration and slow release of incorporated GFs. In vivo, VWF A1 HBD-functionalized fibrin matrices increased angiogenesis and GF retention in VWF-deficient mice. Treatment of chronic skin wounds in diabetic mice with VEGF-A165 and PDGF-BB incorporated within VWF A1 HBD-functionalized fibrin matrices accelerated wound healing, with increased angiogenesis and smooth muscle cell proliferation. Therefore, the VWF A1 HBD can function as a GF reservoir, leading to effective angiogenesis and tissue regeneration.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 114, No. 13 ( 2009-09-24), p. 2819-2828
    Abstract: ADAMTS13 metalloprotease regulates the multimeric size of von Willebrand factor (VWF) by cleaving the Tyr1605-Met1606 bond in the VWF A2 domain. The mechanisms of VWF recognition by ADAMTS13 have yet to be fully resolved. Most studies have focused on the role of exosites within the VWF A2 domain, involved in interaction with the ADAMTS13 spacer domain. In the present study, we expressed different C-terminal domain VWF fragments and evaluated their binding to ADAMTS13 and its truncated mutants, MDTCS and del(TSP5-CUB). Using plate binding assay and surface plasmon resonance, we identified a novel ADAMTS13 binding site (KD ∼ 86 nM) in the region of VWF spanning residues 1874 to 2813, which includes the VWF D4 domain and that interacts with the C-terminal domains of ADAMTS13. We show that the interaction occurs even when VWF is in static conditions, assumed to be globular and where the VWF A2 domain is hidden. We demonstrate that C-terminal VWF fragments, as well as an antibody specifically directed toward the VWF D4 domain, inhibit VWF proteolysis by ADAMTS13 under shear conditions. We propose that this novel VWF C-terminal binding site may participate as the initial step of a multistep interaction ultimately leading to proteolysis of VWF by ADAMTS13.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 115, No. 13 ( 2010-04-01), p. 2666-2673
    Abstract: von Willebrand factor (VWF) multimeric composition is regulated in plasma by ADAMTS13. VWF deglycosylation enhances proteolysis by ADAMTS13. In this study, the role of terminal sialic acid residues on VWF glycans in mediating proteolysis by ADAMTS13 was investigated. Quantification and distribution of VWF sialylation was examined by sequential digestion and high-performance liquid chromatography analysis. Total sialic acid expression on VWF was 167nmol/mg, of which the majority (80.1%) was present on N-linked glycan chains. Enzymatic desialylation of VWF by α2-3,6,8,9 neuraminidase (Neu-VWF) markedly impaired ADAMTS13-mediated VWF proteolysis. Neu-VWF collagen binding activity was reduced to 50% (± 14%) by ADAMTS13, compared with 11% (± 7%) for untreated VWF. Despite this, Neu-VWF exhibited increased susceptibility to other proteases, including trypsin, chymotrypsin, and cathepsin B. VWF expressing different blood groups exhibit altered ADAMTS13 proteolysis rates (O ≥ B 〉 A ≥ AB). However, ABO blood group regulation of ADAMTS13 proteolysis was ablated on VWF desialylation, as both Neu-O-VWF and Neu-AB-VWF were cleaved by ADAMTS13 at identical rates. These novel data show that sialic acid protects VWF against proteolysis by serine and cysteine proteases but specifically enhances susceptibility to ADAMTS13 proteolysis. Quantitative variation in VWF sialylation therefore represents a key determinant of VWF multimeric composition and, as such, may be of pathophysiologic significance.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 2275-2275
    Abstract: Patients with severe haemophilia A (SHA, FVIII & lt;1 IU/dL) and inhibitors to factor VIII, who are resistant to standard immune tolerance (ITI), often have significant morbidity and progressive arthropathy and may be extremely challenging and expensive to manage. Recently, the use of rituximab to treat resistant inhibitors has attracted much interest. Our review of the world literature (July 2008) identified 28 case reports of patients with SHA and inhibitors treated with rituximab. 16 of 28 (57%) achieved an undetectable Bethesda titre. Of the patients who received concomitant FVIII, 15 of 20 (75%) achieved a negative Bethesda titre compared with 1 of 7 (14%) of those treated without FVIII. These data, however, are potentially unreliable because positive outcomes are more likely to be reported and follow up is often short, with relapses unlikely to be reported. We, therefore, collected information on all patients in the UK with SHA treated with rituximab for resistant inhibitors until May 2008. There were 15 patients from 7 centres (9 included in world literature) with a median age of 6 (range 2–37) yrs. There was a disproportionate number of Africans in the cohort compared to the UK population. All had failed at least one previous ITI. Patients were initially treated with 4 weekly doses of rituximab at 375mg/m2, 12 received concomitant FVIII and 4 received additional immunosuppression. One patient had headaches and nausea at the time of the rituximab. Median follow up was 104 (range 30–256) weeks. CR was defined as a negative Bethesda titre, PR as an inhibitor titre & lt;5BU with measurable FVIII recovery or clinical improvement. Age Yrs Ethnicity Mutation Historic peak titre BU/ml No. failed ITI No. Ritux doses FVIII regimen Immuno suppress Time to negative Bethesda Weeks or lowest titre ( ) Relapse Time to relapseWeeks 2nd CR WeeksFollow up Negative Bethesda 3 Cauc C6124T 162 2 4 100/kg/d No 4 Yes 43 No 58 12 Cauc C6496T 125 4 4 To keep FVIII & gt;5IU/dL Vincrist Steroid 4 Yes 156 Yes 204 11 Cauc C1336T 59 1 4 200/kg/d No 44 Yes x2 56 Yes 256 4 Cauc Inv 22 900 1 4 100/kg x3/week Steroid azathio 24 No NA NA 182 10 African Del exon 14 292 2 4 200/kg/d No 84 No NA NA 104 Partial response 3 African Del C5620 369 1 4 200/kg/d No (0.7) No NA NA 84 2 African Inv 22 39 1 4 200/kg/d No (0.5) No NA NA 56 7 African Inv 22 222 1 4 200/kg/d No (1.8) No NA NA 30 4 African G1595A 700 1 4+4 200/kg/d No (1) No NA NA 108 6 Cauc Inv 22 510 2 4+2 200/kg/d Vincrist steroid (1) Yes 2 NA 46 No response 13 African No data 28 1 4 None No NA NA NA NA 104 16 Asian Del intron 18–21 & gt;4000 2 4 None No NA NA NA NA 156 6 Cauc Frame shift C1876 780 1 4 200/kg/d Steroid NA NA NA NA 156 2 African Inv 22 6300 1 4 200/kg/d No NA NA NA NA 56 37 Cauc No data 780 1 4 None No NA NA NA NA 104 A negative Bethesda titre was achieved in 5 patients (33%) and was sustained in 2 (13%). Of the 3 who relapsed, 2 achieved a second negative Bethesda after further treatment (one with FVIII and one with FVIII + rituximab). The other did not respond to further rituximab without FVIII. Of the 5 patients with a PR one relapsed but 4 continue to be managed on FVIII with prolonged follow up and without significant bleeding. They appear to have been converted from high to low responders. Therefore, 8 of 15 (53%) of patients have had a prolonged benefit from rituximab treatment. There were 5 non responders, including all 3 who did not receive concomitant FVIII. Therefore, 8 of 12 (67%) patients who received FVIII had a clinical benefit. These data are a consecutive national cohort with prolonged follow up and are therefore less likely to be affected by reporting bias. The results are inferior to the published literature but demonstrate that, in patients with SHA and inhibitors resistant to standard ITI, rituximab in association with FVIII leads to sustained clinical improvement in a significant proportion of patients, although complete eradication of the inhibitor is uncommon.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...