Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Am J Physiol Heart Circ Physiol ; 322(1): H87-H93, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34890277

RESUMO

The 2019 coronavirus disease (COVID-19) is the disease caused by SARS-CoV-2 infection. Although this infection has been shown to affect the respiratory system, a high incidence of thrombotic events has been observed in severe cases of COVID-19 and in a significant portion of COVID-19 nonsurvivors. Although prior literature has reported on both the coagulopathy and hypercoagulability of COVID-19, the specifics of coagulation have not been fully investigated. Observations of microthrombosis in patients with COVID-19 have brought attention to potential inflammatory endothelial injury. Von Willebrand factor (VWF) and its protease, A disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13 (ADAMTS13), play an important homeostatic role in responding to endothelial injury. This report provides an overview of the literature investigating the role the VWF/ADAMTS13 axis may have in COVID-19 thrombotic events and suggests potential therapeutic strategies to prevent the progression of coagulopathy in patients with COVID-19.


Assuntos
Proteína ADAMTS13/metabolismo , Transtornos da Coagulação Sanguínea/metabolismo , COVID-19/sangue , Fator de von Willebrand/metabolismo , Coagulação Sanguínea , Transtornos da Coagulação Sanguínea/etiologia , COVID-19/complicações , Humanos
2.
Haematologica ; 107(3): 668-679, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33763999

RESUMO

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.


Assuntos
Galactose , Ácido N-Acetilneuramínico , Fator de von Willebrand , Galactose/metabolismo , Humanos , Lectinas/metabolismo , Macrófagos/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Polissacarídeos/metabolismo , Fator de von Willebrand/metabolismo
3.
Blood ; 143(19): 1887-1888, 2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38722656
4.
Blood ; 133(24): 2559-2569, 2019 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-30975637

RESUMO

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.


Assuntos
Neovascularização Fisiológica/fisiologia , Cicatrização/fisiologia , Fator de von Willebrand/metabolismo , Animais , Diabetes Mellitus Experimental , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Domínios Proteicos
5.
Blood ; 141(10): 1102-1103, 2023 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-36893008
6.
Blood ; 137(8): 1007-1008, 2021 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-33630048
7.
Blood ; 128(15): 1959-1968, 2016 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-27554083

RESUMO

Enhanced von Willebrand factor (VWF) clearance is important in the etiology of von Willebrand disease. However, the molecular mechanisms underlying VWF clearance remain poorly understood. In this study, we investigated the role of VWF domains and specific glycan moieties in regulating in vivo clearance. Our findings demonstrate that the A1 domain of VWF contains a receptor-recognition site that plays a key role in regulating the interaction of VWF with macrophages. In A1-A2-A3 and full-length VWF, this macrophage-binding site is cryptic but becomes exposed following exposure to shear or ristocetin. Previous studies have demonstrated that the N-linked glycans within the A2 domain play an important role in modulating susceptibility to ADAMTS13 proteolysis. We further demonstrate that these glycans presented at N1515 and N1574 also play a critical role in protecting VWF against macrophage binding and clearance. Indeed, loss of the N-glycan at N1515 resulted in markedly enhanced VWF clearance that was significantly faster than that observed with any previously described VWF mutations. In addition, A1-A2-A3 fragments containing the N1515Q or N1574Q substitutions also demonstrated significantly enhanced clearance. Importantly, clodronate-induced macrophage depletion significantly attenuated the increased clearance observed with N1515Q and N1574Q in both full-length VWF and A1-A2-A3. Finally, we further demonstrate that loss of these N-linked glycans does not enhance clearance in VWF in the presence of a structurally constrained A2 domain. Collectively, these novel findings support the hypothesis that conformation of the VWF A domains plays a critical role in modulating macrophage-mediated clearance of VWF in vivo.


Assuntos
Macrófagos/metabolismo , Polissacarídeos/metabolismo , Fator de von Willebrand/metabolismo , Substituição de Aminoácidos , Animais , Linhagem Celular Tumoral , Humanos , Macrófagos/citologia , Camundongos , Camundongos Knockout , Mutação de Sentido Incorreto , Polissacarídeos/química , Polissacarídeos/genética , Domínios Proteicos , Fator de von Willebrand/química , Fator de von Willebrand/genética
8.
Blood ; 121(14): 2773-84, 2013 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-23355534

RESUMO

Von Willebrand disease (VWD) is a heterogeneous bleeding disorder caused by decrease or dysfunction of von Willebrand factor (VWF). A wide range of mutations in the VWF gene have been characterized; however, their cellular consequences are still poorly understood. Here we have used a recently developed approach to study the molecular and cellular basis of VWD. We isolated blood outgrowth endothelial cells (BOECs) from peripheral blood of 4 type 1 VWD and 4 type 2 VWD patients and 9 healthy controls. We confirmed the endothelial lineage of BOECs, then measured VWF messenger RNA (mRNA) and protein levels (before and after stimulation) and VWF multimers. Decreased mRNA levels were predictive of plasma VWF levels in type 1 VWD, confirming a defect in VWF synthesis. However, BOECs from this group of patients also showed defects in processing, storage, and/or secretion of VWF. Levels of VWF mRNA and protein were normal in BOECs from 3 type 2 VWD patients, supporting the dysfunctional VWF model. However, 1 type 2M patient showed decreased VWF synthesis and storage, indicating a complex cellular defect. These results demonstrate for the first time that isolation of endothelial cells from VWD patients provides novel insight into cellular mechanisms of the disease.


Assuntos
Células Endoteliais/citologia , Células Endoteliais/fisiologia , Doença de von Willebrand Tipo 1 , Doença de von Willebrand Tipo 2 , Fator de von Willebrand/genética , Adulto , Idoso , Linhagem da Célula/fisiologia , Células Cultivadas , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fenótipo , RNA Mensageiro/metabolismo , Corpos de Weibel-Palade/metabolismo , Doença de von Willebrand Tipo 1/genética , Doença de von Willebrand Tipo 1/metabolismo , Doença de von Willebrand Tipo 1/patologia , Doença de von Willebrand Tipo 2/genética , Doença de von Willebrand Tipo 2/metabolismo , Doença de von Willebrand Tipo 2/patologia , Fator de von Willebrand/metabolismo
9.
Blood ; 120(1): 214-22, 2012 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-22517896

RESUMO

We have examined the effect of the O-linked glycan (OLG) structures of VWF on its interaction with the platelet receptor glycoprotein Ibα. The 10 OLGs were mutated individually and as clusters (Clus) on either and both sides of the A1 domain: Clus1 (N-terminal side), Clus2 (C-terminal side), and double cluster (DC), in both full-length-VWF and in a VWF construct spanning D' to A3 domains. Mutations did not alter VWF secretion by HEK293T cells, multimeric structure, or static collagen binding. The T1255A, Clus1, and DC variants caused increased ristocetin-mediated GPIbα binding to VWF. Platelet translocation rate on OLG mutants was increased because of reduced numbers of GPIbα binding sites but without effect on bond lifetime. In contrast, OLG mutants mediated increased platelet capture on collagen under high shear stress that was associated with increased adhesion of these variants to the collagen under flow. These findings suggest that removal of OLGs increases the flexibility of the hinge linker region between the D3 and A1 domain, facilitating VWF unfolding by shear stress, thereby enhancing its ability to bind collagen and capture platelets. These data demonstrate an important functional role of VWF OLGs under shear stress conditions.


Assuntos
Plaquetas/fisiologia , Glicoproteínas de Membrana/metabolismo , Polissacarídeos/metabolismo , Fator de von Willebrand/genética , Fator de von Willebrand/metabolismo , Antibacterianos/farmacologia , Sítios de Ligação/fisiologia , Colágeno/metabolismo , Variação Genética , Glicosilação , Células HEK293 , Humanos , Glicoproteínas de Membrana/química , Mutagênese/fisiologia , Complexo Glicoproteico GPIb-IX de Plaquetas , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína/fisiologia , Proteínas Recombinantes/metabolismo , Fluxo Sanguíneo Regional/fisiologia , Ristocetina/farmacologia , Estresse Mecânico , Fator de von Willebrand/química
10.
Blood ; 120(25): 5059-62, 2012 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-23074276

RESUMO

Protein S is a cofactor for tissue factor pathway inhibitor (TFPI) that critically reduces the inhibition constant for FXa to below the plasma concentration of TFPI. TFPI Kunitz domain 3 is required for this enhancement to occur. To delineate the molecular mechanism underlying enhancement of TFPI function, in the present study, we produced a panel of Kunitz domain 3 variants of TFPI encompassing all 12 surface-exposed charged residues. Thrombin-generation assays in TFPI-depleted plasma identified a novel variant, TFPI E226Q, which exhibited minimal enhancement by protein S. This was confirmed in purified FXa inhibition assays in which no protein S enhancement of TFPI E226Q was detected. Surface plasmon resonance demonstrated concentration-dependent binding of protein S to wild-type TFPI, but almost no binding to TFPI E226Q. We conclude that the TFPI Kunitz domain 3 residue Glu226 is essential for TFPI enhancement by protein S.


Assuntos
Lipoproteínas/genética , Lipoproteínas/metabolismo , Mutação Puntual , Proteína S/metabolismo , Fator Xa/metabolismo , Inibidores do Fator Xa , Humanos , Lipoproteínas/química , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ressonância de Plasmônio de Superfície , Trombina/metabolismo
11.
J Thromb Haemost ; 22(7): 1867-1879, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38608731

RESUMO

BACKGROUND: Carfilzomib (CFZ) is a second-generation proteasome inhibitor used to treat multiple myeloma. Potent inhibition of the proteasome results in chronic proteotoxic endoplasmic reticulum (ER) stress, leading to apoptosis. While CFZ has improved survival rates in multiple myeloma, it is associated with an increased risk of cardiovascular adverse effects. While this has been putatively linked to cardiotoxicity, CFZ could potentially also exhibit adverse effects on the endothelium. OBJECTIVES: To investigate the effects of CFZ on the endothelium. METHODS: Human umbilical vein endothelial cells (HUVECs) were treated with CFZ, and expression of relevant markers of ER stress, inflammation, and thrombosis was measured and functionally assessed. RESULTS: CFZ failed to induce ER stress in HUVECs but induced the expression of Kruppel-like factor 4, endothelial nitric oxide synthase, tissue plasminogen activator, and thrombomodulin and reduced tumor necrosis factor alpha (TNFα)-mediated intercellular adhesion molecule 1 and tissue factor expression, suggesting a potential protective effect on the endothelium. Consistent with these observations, CFZ reduced leukocyte adhesion under shear stress and reduced factor Xa generation and fibrin clot formation on the endothelium following TNFα treatment and inhibited von Willebrand factor (VWF) and angiopoietin-2 exocytosis from Weibel-Palade bodies. Subsequently, CFZ inhibited the formation of VWF-platelet strings, and moreover, media derived from myeloma cell lines induced VWF release, a process also inhibited by CFZ. CONCLUSION: These data demonstrate that CFZ is unable to induce ER stress in confluent resting endothelial cells and can conversely attenuate the prothrombotic effects of TNFα on the endothelium. This study suggests that CFZ does not negatively alter HUVECs, and proteasome inhibition of the endothelium may offer a potential way to prevent thrombosis.


Assuntos
Anti-Inflamatórios , Estresse do Retículo Endoplasmático , Fibrinolíticos , Células Endoteliais da Veia Umbilical Humana , Oligopeptídeos , Inibidores de Proteassoma , Humanos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Oligopeptídeos/farmacologia , Inibidores de Proteassoma/farmacologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Fibrinolíticos/farmacologia , Anti-Inflamatórios/farmacologia , Adesão Celular/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Tromboplastina/metabolismo , Leucócitos/efeitos dos fármacos , Leucócitos/metabolismo , Trombose/prevenção & controle , Trombose/induzido quimicamente , Trombose/metabolismo , Células Cultivadas , Inflamação/metabolismo , Trombomodulina
12.
Blood ; 117(3): 1071-80, 2011 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-21048155

RESUMO

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 αvß3 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.


Assuntos
Células Endoteliais/metabolismo , Neovascularização Fisiológica , Fator de von Willebrand/metabolismo , Adulto , Idoso de 80 Anos ou mais , Angiopoietina-2/genética , Angiopoietina-2/metabolismo , Animais , Linhagem Celular , Movimento Celular , Proliferação de Células , Células Endoteliais/citologia , Feminino , Hemostasia , Humanos , Immunoblotting , Integrina alfaVbeta3/genética , Integrina alfaVbeta3/metabolismo , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Neovascularização Patológica , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Doenças de von Willebrand/genética , Doenças de von Willebrand/metabolismo , Doenças de von Willebrand/patologia , Fator de von Willebrand/genética
13.
Biochem J ; 447(2): 217-28, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22849435

RESUMO

vWF (von Willebrand factor) is a key component for maintenance of normal haemostasis, acting as the carrier protein of the coagulant Factor VIII and mediating platelet adhesion at sites of vascular injury. There is ample evidence that vWF glycan moieties are crucial determinants of its expression and function. Of particular clinical interest, ABH antigens influence vWF plasma levels according to the blood group of individuals, although the molecular mechanism underlying this phenomenon remains incompletely understood. The present paper reports analyses of the human plasma vWF N-glycan population using advanced MS. Glycomics analyses revealed approximately 100 distinct N-glycan compositions and identified a variety of structural features, including lactosaminic extensions, ABH antigens and sulfated antennae, as well as bisecting and terminal GlcNAc residues. We estimate that some 300 N-glycan structures are carried by human vWF. Glycoproteomics analyses mapped ten of the consensus sites known to carry N-glycans. Glycan populations were found to be distinct, although many structural features were shared across all sites. Notably, the H antigen is not restricted to particular N-glycosylation sites. Also, the Asn(2635) site, previously designated as unoccupied, was found to be highly glycosylated. The delineation of such varied glycan populations in conjunction with current models explaining vWF activity will facilitate research aimed at providing a better understanding of the influence of glycosylation on vWF function.


Assuntos
Glicômica/métodos , Fator de von Willebrand/química , Sequência de Aminoácidos , Antígenos de Bactérias/química , Sequência de Carboidratos , Glicosilação , Humanos , Dados de Sequência Molecular , Polissacarídeos/química , Proteômica/métodos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Espectrometria de Massas em Tandem
14.
J Thromb Haemost ; 21(7): 1802-1812, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37011710

RESUMO

BACKGROUND: The von Willebrand factor (VWF) is a multimeric plasma glycoprotein essential for hemostasis, inflammation, and angiogenesis. The majority of VWF is synthesized by endothelial cells (ECs) and stored in Weibel-Palade bodies (WPB). Among the range of proteins shown to co-localize to WPB is angiopoietin-2 (Angpt-2), a ligand of the receptor tyrosine kinase Tie-2. We have previously shown that VWF itself regulates angiogenesis, raising the hypothesis that some of the angiogenic activity of VWF may be mediated by its interaction with Angpt-2. METHODS: Static-binding assays were used to probe the interaction between Angpt-2 and VWF. Binding in media from cultured human umbilical vein ECs s and in plasma was determined by immunoprecipitation experiments. Immunofluorescence was used to detect the presence of Angpt-2 on VWF strings, and flow assays were used to investigate the effect on VWF function. RESULTS: Static-binding assays revealed that Angpt-2 bound to VWF with high affinity (KD,app ∼3 nM) in a pH and calcium-dependent manner. The interaction was localized to the VWF A1 domain. Co-immunoprecipitation experiments demonstrated that the complex persisted following stimulated secretion from ECs and was present in plasma. Angpt-2 was also visible on VWF strings on stimulated ECs. The VWF-Angpt-2 complex did not inhibit the binding of Angpt-2 to Tie-2 and did not significantly interfere with VWF-platelet capture. CONCLUSIONS: Together, these data demonstrate a direct binding interaction between Angpt-2 and VWF that persists after secretion. VWF may act to localize Angpt-2; further work is required to establish the functional consequences of this interaction.


Assuntos
Corpos de Weibel-Palade , Fator de von Willebrand , Humanos , Fator de von Willebrand/metabolismo , Corpos de Weibel-Palade/metabolismo , Angiopoietina-2/metabolismo , Exocitose , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Cultivadas
15.
Blood ; 116(4): 640-8, 2010 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-20418283

RESUMO

We examined the role that N-linked glycans play in the synthesis and expression of von Willebrand Factor (VWF). Blocking the addition of N-linked glycans (NLGs) or inhibiting initial glycan processing prevented secretion of VWF. To determine whether specific glycosylation sites were important, the 16 VWF N-linked glycosylation sites were mutated followed by expression in HEK293T cells. Four NLG mutants affected VWF expression: N99Q (D1 domain), N857Q (D' domain), N2400Q (B1 domain), and N2790Q (CK domain) either abolished or reduced secretion of VWF and this was confirmed by metabolic labeling. Multimer analysis of mutant N2790Q cell lysate revealed an increase in VWF monomers, which was also observed when the isolated CK domain was expressed with N2790 mutated. Immunofluorescence microscopy showed that mutants N99Q, N857Q, and N2790Q were primarily retained within the ER, producing only few pseudo Weibel-Palade bodies over longer time periods compared with wtVWF. All the variants also showed an increase in free thiol reactivity. This was greatest with N857Q and D4-C2 NLG mutants, which had approximately 6-fold and 3- to 4-fold more free thiol reactivity than wtVWF. These data provide further evidence of the critical role that individual N-linked glycans play in determining VWF synthesis and expression.


Assuntos
Fator de von Willebrand/biossíntese , Fator de von Willebrand/metabolismo , Substituição de Aminoácidos/fisiologia , Antígenos de Bactérias/química , Antígenos de Bactérias/metabolismo , Domínio Catalítico/genética , Células Cultivadas , Expressão Gênica , Glicosilação , Humanos , Modelos Biológicos , Proteínas Mutantes/metabolismo , Mutação/fisiologia , Polissacarídeos/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Transporte Proteico , Especificidade por Substrato , Distribuição Tecidual , Fator de von Willebrand/química , Fator de von Willebrand/genética
16.
Blood ; 115(13): 2666-73, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19965639

RESUMO

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 alpha2-3,6,8,9 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 > or = B > A > or = 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.


Assuntos
Proteínas ADAM/metabolismo , Ácido N-Acetilneuramínico/fisiologia , Fator de von Willebrand/química , Sistema ABO de Grupos Sanguíneos/química , Sistema ABO de Grupos Sanguíneos/metabolismo , Proteína ADAMTS13 , Biopolímeros , Configuração de Carboidratos , Colágeno/metabolismo , Cisteína Proteases/metabolismo , Galactose/química , Glicosídeo Hidrolases/farmacologia , Humanos , Ácido N-Acetilneuramínico/química , Neuraminidase/farmacologia , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase/farmacologia , Conformação Proteica , Processamento de Proteína Pós-Traducional , Serina Proteases/metabolismo , Especificidade por Substrato , alfa-N-Acetilgalactosaminidase/farmacologia , Fator de von Willebrand/efeitos dos fármacos , Fator de von Willebrand/metabolismo
17.
Blood ; 114(16): 3489-96, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19687512

RESUMO

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.


Assuntos
Colágeno Tipo II/metabolismo , Colágeno Tipo I/metabolismo , Hemorragia/metabolismo , Mutação de Sentido Incorreto , Multimerização Proteica/genética , Doenças de von Willebrand/metabolismo , Fator de von Willebrand/metabolismo , Substituição de Aminoácidos , Sítios de Ligação/genética , Linhagem Celular , Colágeno Tipo I/genética , Colágeno Tipo II/genética , Família , Feminino , Expressão Gênica , Hemorragia/genética , Humanos , Masculino , Mutagênese Sítio-Dirigida , Ligação Proteica/genética , Estrutura Terciária de Proteína/genética , Proteínas Recombinantes/economia , Proteínas Recombinantes/metabolismo , Doenças de von Willebrand/classificação , Doenças de von Willebrand/genética , Fator de von Willebrand/genética
18.
Blood ; 114(13): 2819-28, 2009 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-19587373

RESUMO

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 (K(D) approximately 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.


Assuntos
Proteínas ADAM/metabolismo , Dobramento de Proteína , Fator de von Willebrand/química , Fator de von Willebrand/metabolismo , Proteínas ADAM/química , Proteína ADAMTS13 , Sítios de Ligação , Células Cultivadas , Humanos , Modelos Biológicos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Mapeamento de Interação de Proteínas , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína/fisiologia , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
19.
J Thromb Haemost ; 19(2): 358-369, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33075181

RESUMO

BACKGROUND: Von Willebrand factor (VWF) contains a number of free thiols, the majority of which are located in its C-domains, and these have been shown to alter VWF function, However, the impact of free thiols on function following acute exposure of VWF to collagen under high and pathological shear stress has not been determined. METHODS: VWF free thiols were blocked with N-ethylmaleimide and flow assays performed under high and pathological shear rates to determine the impact on platelet capture and collagen binding function. Atomic force microscopy (AFM) was used to probe the interaction of VWF with collagen and molecular simulations conducted to determine the effect of free thiols on the flexibility of the VWF-C4 domain. RESULTS: Blockade of VWF free thiols reduced VWF-mediated platelet capture to collagen in a shear-dependent manner, with platelet capture virtually abolished above 5000 s-1 and in regions of stenosis in microfluidic channels. Direct visualization of VWF fibers formed under extreme pathological shear rates and analysis of collagen-bound VWF attributed the effect to altered binding of VWF to collagen. AFM measurements showed that thiol-blockade reduced the lifetime and strength of the VWF-collagen bond. Pulling simulations of the VWF-C4 domain demonstrated that with one or two reduced disulphide bonds the C4 domain has increased flexibility and the propensity to undergo free-thiol exchange. CONCLUSIONS: We conclude that free thiols in the C-domains of VWF enhance the flexibility of the molecule and enable it to withstand high shear forces following collagen binding, demonstrating a previously unrecognized role for VWF free thiols.


Assuntos
Compostos de Sulfidrila , Fator de von Willebrand , Plaquetas/metabolismo , Colágeno/metabolismo , Humanos , Adesividade Plaquetária , Ligação Proteica , Estresse Mecânico , Fator de von Willebrand/metabolismo
20.
Blood ; 111(6): 3042-9, 2008 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17975018

RESUMO

We examined the role of N-linked glycan structures of VWF on its interaction with ADAMTS13. PNGase F digestion followed by lectin analysis demonstrated that more than 90% of VWF N-linked glycan chains could be removed from the molecule (PNG-VWF) without disruption of its multimeric structure or its ability to bind to collagen. PNG-VWF had an approximately 4-fold increased affinity for ADAMTS13 compared with control VWF. PNG-VWF was cleaved by ADAMTS13 faster than control VWF and was also proteolysed in the absence of urea. Occupancy of the N-linked glycan sites at N1515 and N1574 and their presentation of ABO(H) blood group sugars were confirmed with an isolated tryptic fragment. Recombinant VWF was mutated to prevent glycosylation at these sites. Mutation of N1515 did not alter ADAMTS13 binding or increase rate of ADAMTS13 proteolysis. Mutation of N1574 increased the susceptibility of VWF to ADAMTS13 proteolysis and allowed cleavage in the absence of urea. Mutation of N1574 in the isolated recombinant VWF-A2 domain also increased binding and ADAMTS13 proteolysis. These data demonstrate that the N-linked glycans of VWF have a modulatory effect on the interaction with ADAMTS13. At least part of this effect is conformational, but steric hindrance may also be important.


Assuntos
Proteínas ADAM/metabolismo , Fator de von Willebrand/metabolismo , Proteínas ADAM/genética , Linhagem Celular , Colágeno/metabolismo , Glicosilação , Humanos , Modelos Moleculares , Mutação/genética , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Fator de von Willebrand/química , Fator de von Willebrand/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA