Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Eur Respir J ; 53(3)2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30655285

RESUMEN

Chronic thromboembolic pulmonary hypertension (CTEPH) is an important consequence of pulmonary embolism that is associated with abnormalities in haemostasis. We investigated the ADAMTS13-von Willebrand factor (VWF) axis in CTEPH, including its relationship with disease severity, inflammation, ABO groups and ADAMTS13 genetic variants.ADAMTS13 and VWF plasma antigen levels were measured in patients with CTEPH (n=208), chronic thromboembolic disease without pulmonary hypertension (CTED) (n=35), resolved pulmonary embolism (n=28), idiopathic pulmonary arterial hypertension (n=30) and healthy controls (n=68). CTEPH genetic ABO associations and protein quantitative trait loci were investigated. ADAMTS13-VWF axis abnormalities were assessed in CTEPH and healthy control subsets by measuring ADAMTS13 activity, D-dimers and VWF multimeric size.Patients with CTEPH had decreased ADAMTS13 (adjusted ß -23.4%, 95% CI -30.9- -15.1%, p<0.001) and increased VWF levels (ß +75.5%, 95% CI 44.8-113%, p<0.001) compared to healthy controls. ADAMTS13 levels remained low after reversal of pulmonary hypertension by pulmonary endarterectomy surgery and were equally reduced in CTED. We identified a genetic variant near the ADAMTS13 gene associated with ADAMTS13 protein that accounted for ∼8% of the variation in levels.The ADAMTS13-VWF axis is dysregulated in CTEPH. This is unrelated to pulmonary hypertension, disease severity or markers of systemic inflammation and implicates the ADAMTS13-VWF axis in CTEPH pathobiology.


Asunto(s)
Proteína ADAMTS13/genética , Hipertensión Pulmonar/fisiopatología , Embolia Pulmonar/fisiopatología , Factor de von Willebrand/análisis , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores , Estudios de Casos y Controles , Enfermedad Crónica , Endarterectomía , Femenino , Humanos , Hipertensión Pulmonar/genética , Modelos Lineales , Masculino , Persona de Mediana Edad , Análisis Multivariante , Embolia Pulmonar/genética , Trombosis/genética , Trombosis/fisiopatología
2.
J Biol Chem ; 292(14): 5760-5769, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-28209710

RESUMEN

Blood loss is prevented by the multidomain glycoprotein von Willebrand factor (VWF), which binds exposed collagen at damaged vessels and captures platelets. VWF is regulated by the metalloprotease ADAMTS13, which in turn is conformationally activated by VWF. To delineate the structural requirements for VWF-mediated conformational activation of ADAMTS13, we performed binding and functional studies with a panel of truncated ADAMTS13 variants. We demonstrate that both the isolated CUB1 and CUB2 domains in ADAMTS13 bind to the spacer domain exosite of a truncated ADAMTS13 variant, MDTCS (KD of 135 ± 1 0.1 nm and 86.9 ± 9.0 nm, respectively). However, only the CUB1 domain inhibited proteolytic activity of MDTCS. Moreover, ADAMTS13ΔCUB2, unlike ADAMTS13ΔCUB1-2, exhibited activity similar to wild-type ADAMTS13 and could be activated by VWF D4-CK. The CUB2 domain is, therefore, not essential for maintaining the inactive conformation of ADAMTS13. Both CUB domains could bind to the VWF D4-CK domain fragment (KD of 53.7 ± 2.1 nm and 84.3 ± 2.0 nm, respectively). However, deletion of both CUB domains did not prevent VWF D4-CK binding, suggesting that competition for CUB-domain binding to the spacer domain is not the dominant mechanism behind the conformational activation. ADAMTS13ΔTSP8-CUB2 could no longer bind to VWF D4-CK, and deletion of TSP8 abrogated ADAMTS13 conformational activation. These findings support an ADAMTS13 activation model in which VWF D4-CK engages the TSP8-CUB2 domains, inducing the conformational change that disrupts the CUB1-spacer domain interaction and thereby activates ADAMTS13.


Asunto(s)
Proteína ADAMTS13/química , Modelos Químicos , Factor de von Willebrand/química , Proteína ADAMTS13/metabolismo , Células HEK293 , Humanos , Unión Proteica/fisiología , Dominios Proteicos , Factor de von Willebrand/metabolismo
3.
J Biol Chem ; 292(22): 9335-9344, 2017 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-28420729

RESUMEN

Tissue factor pathway inhibitor (TFPI), the main inhibitor of initiation of coagulation, exerts an important anticoagulant role through the factor Xa (FXa)-dependent inhibition of tissue factor/factor VIIa. Protein S is a TFPI cofactor, enhancing the efficiency of FXa inhibition. TFPI can also inhibit prothrombinase assembly by directly interacting with coagulation factor V (FV), which has been activated by FXa. Because full-length TFPI associates with FV in plasma, we hypothesized that FV may influence TFPI inhibitory function. Using pure component FXa inhibition assays, we found that although FV alone did not influence TFPI-mediated FXa inhibition, it further enhanced TFPI in the presence of protein S, resulting in an ∼8-fold reduction in Ki compared with TFPI alone. A FV variant (R709Q/R1018Q/R1545Q, FVΔIIa) that cannot be cleaved/activated by thrombin or FXa also enhanced TFPI-mediated inhibition of FXa ∼12-fold in the presence of protein S. In contrast, neither activated FV nor recombinant B-domain-deleted FV could enhance TFPI-mediated inhibition of FXa in the presence of protein S, suggesting a functional contribution of the B domain. Using TFPI and protein S variants, we show further that the enhancement of TFPI-mediated FXa inhibition by protein S and FV depends on a direct protein S/TFPI interaction and that the TFPI C-terminal tail is not essential for this enhancement. In FXa-catalyzed prothrombin activation assays, both FV and FVΔIIa (but not activated FV) enhanced TFPI function in the presence of protein S. These results demonstrate a new anticoagulant (cofactor) function of FV that targets the early phase of coagulation before prothrombinase assembly.


Asunto(s)
Anticoagulantes/metabolismo , Coagulación Sanguínea/fisiología , Factor V/metabolismo , Sustitución de Aminoácidos , Factor V/genética , Factor Xa/genética , Factor Xa/metabolismo , Humanos , Lipoproteínas/genética , Lipoproteínas/metabolismo , Mutación Missense , Dominios Proteicos , Proteína S/genética , Proteína S/metabolismo , Protrombina/genética , Protrombina/metabolismo
4.
Blood ; 127(13): 1711-8, 2016 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-26773038

RESUMEN

Shear forces in the blood trigger a conformational transition in the von Willebrand factor (VWF) A2 domain, from its native folded to an unfolded state, in which the cryptic scissile bond (Y1605-M1606) is exposed and can then be proteolysed by ADAMTS13. The conformational transition depends upon a Ca(2+)binding site and a vicinal cysteine disulfide bond. Glycosylation at N1574 has previously been suggested to modulate VWF A2 domain interaction with ADAMTS13 through steric hindrance by the bulky carbohydrate structure. We investigated how the N-linked glycans of the VWF A2 domain affect thermostability and regulate both the exposure of the ADAMTS13 binding sites and the scissile bond. We show by differential scanning fluorimetry that the N-linked glycans thermodynamically stabilize the VWF A2 domain. The essential component of the glycan structure is the first sugar residue (GlcNAc) at the N1574 attachment site. From its crystal structures, N1574-GlcNAc is predicted to form stabilizing intradomain interactions with Y1544 and nearby residues. Substitution of the surface-exposed Y1544 to aspartic acid is able to stabilize the domain in the absence of glycosylation and protect against ADAMTS13 proteolysis in both the VWF A2 domain and FLVWF. Glycan stabilization of the VWF A2 domain acts together with the Ca(2+)binding site and vicinal cysteine disulfide bond to control unfolding and ADAMTS13 proteolysis.


Asunto(s)
Proteínas ADAM/metabolismo , Polisacáridos/metabolismo , Dominios y Motivos de Interacción de Proteínas , Factor de von Willebrand/química , Factor de von Willebrand/metabolismo , Proteínas ADAM/química , Proteína ADAMTS13 , Acetilglucosamina/química , Acetilglucosamina/metabolismo , Sitios de Unión , Calcio/metabolismo , Cristalografía por Rayos X , Cisteína/química , Cisteína/metabolismo , Células HEK293 , Humanos , Modelos Moleculares , Unión Proteica , Pliegue de Proteína , Dominios y Motivos de Interacción de Proteínas/genética , Estabilidad Proteica , Proteolisis , Factor de von Willebrand/genética
5.
Blood ; 125(12): 1968-75, 2015 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-25564400

RESUMEN

ADAMTS13 proteolytically regulates the platelet-tethering function of von Willebrand factor (VWF). ADAMTS13 function is dependent upon multiple exosites that specifically bind the unraveled VWF A2 domain and enable proteolysis. We carried out a comprehensive functional analysis of the ADAMTS13 cysteine-rich (Cys-rich) domain using engineered glycans, sequence swaps, and single point mutations in this domain. Mutagenesis of Cys-rich domain-charged residues had no major effect on ADAMTS13 function, and 5 out of 6 engineered glycans on the Cys-rich domain also had no effect on ADAMTS13 function. However, a glycan attached at position 476 appreciably reduced both VWF binding and proteolysis. Substitution of Cys-rich sequences for the corresponding regions in ADAMTS1 identified a hydrophobic pocket involving residues Gly471-Val474 as being of critical importance for both VWF binding and proteolysis. Substitution of hydrophobic VWF A2 domain residues to serine in a region (residues 1642-1659) previously postulated to interact with the Cys-rich domain revealed the functional importance of VWF residues Ile1642, Trp1644, Ile1649, Leu1650, and Ile1651. Furthermore, the functional deficit of the ADAMTS13 Cys-rich Gly471-Val474 variant was dependent on these same hydrophobic VWF residues, suggesting that these regions form complementary binding sites that directly interact to enhance the efficiency of the proteolytic reaction.


Asunto(s)
Proteínas ADAM/fisiología , Factor de von Willebrand/química , Proteínas ADAM/química , Proteína ADAMTS13 , Secuencia de Aminoácidos , Sitios de Unión , Cisteína/química , Humanos , Datos de Secuencia Molecular , Mutagénesis , Mutación Puntual , Polisacáridos/química , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Especificidad por Sustrato
6.
Opt Express ; 25(7): 7220-7229, 2017 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-28380847

RESUMEN

We generate coherent ultraviolet radiation at 313 nm as the third harmonic of an external-cavity diode laser. We use this radiation for laser cooling of trapped beryllium atomic ions and sympathetic cooling of co-trapped beryllium-hydride molecular ions. An LBO crystal in an enhancement cavity generates the second harmonic, and a BBO crystal in a doubly resonant enhancement cavity mixes this second harmonic with the fundamental to produce the third harmonic. Each enhancement cavity is preceded by a tapered amplifier to increase the fundamental light. The 36-mW output power of this all-semiconductor-gain system will enable quantum control of the beryllium ions' motion.

7.
Proc Natl Acad Sci U S A ; 111(52): 18578-83, 2014 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-25512499

RESUMEN

A disintegrin and metalloprotease with thrombospondin motifs 13 (ADAMTS13) is a metalloprotease that regulates von Willebrand factor (VWF) function. ADAMTS13-mediated proteolysis is determined by conformational changes in VWF, but also may depend on its own conformational activation. Kinetic analysis of WT ADAMTS13 revealed ∼ 2.5-fold reduced activity compared with ADAMTS13 lacking its C-terminal tail (MDTCS) or its CUB1-2 domains (WTΔCUB1-2), suggesting that the CUB domains naturally limit ADAMTS13 function. Consistent with this suggestion, WT ADAMTS13 activity was enhanced ∼ 2.5-fold by preincubation with either an anti-CUB mAb (20E9) or VWF D4CK (the natural binding partner for the CUB domains). Furthermore, the isolated CUB1-2 domains not only bound MDTCS, but also inhibited activity by up to 2.5-fold. Interestingly, a gain-of-function (GoF) ADAMTS13 spacer domain variant (R568K/F592Y/R660K/Y661F/Y665F) was ∼ 2.5-fold more active than WT ADAMTS13, but could not be further activated by 20E9 mAb or VWF D4CK and was unable to bind or to be inhibited by the CUB1-2 domains, suggesting that the inhibitory effects of the CUB domains involve an interaction with the spacer domain that is disrupted in GoF ADAMTS13. Electron microscopy demonstrated a "closed" conformation of WT ADAMTS13 and suggested a more "open" conformation for GoF ADAMTS13. The cryptic spacer domain epitope revealed by conformational unfolding also represents the core antigenic target for autoantibodies in thrombotic thrombocytopenic purpura. We propose that ADAMTS13 circulates in a closed conformation, which is maintained by a CUB-spacer domain binding interaction. ADAMTS13 becomes conformationally activated on demand through interaction of its C-terminal CUB domains with VWF, making it susceptible to immune recognition.


Asunto(s)
Proteínas ADAM/química , Proteínas ADAM/sangre , Proteínas ADAM/genética , Proteína ADAMTS13 , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Anticuerpos Monoclonales de Origen Murino/química , Activación Enzimática , Humanos , Mutación Missense , Estructura Terciaria de Proteína , Púrpura Trombocitopénica Trombótica/enzimología , Púrpura Trombocitopénica Trombótica/genética , Eliminación de Secuencia , Factor de von Willebrand/química , Factor de von Willebrand/genética , Factor de von Willebrand/metabolismo
8.
Blood ; 123(16): 2585-92, 2014 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-24558203

RESUMEN

Rheological shear forces in the blood trigger von Willebrand factor (VWF) unfolding which exposes the Y1605-M1606 scissile bond within the VWF A2 domain for cleavage by ADAMTS13. The VWF A2 domain contains 2 structural features that provide it with stability: a vicinal disulphide bond and a Ca(2+)-binding site (CBS). We investigated how these 2 structural features interplay to determine stability and regulate the exposure of the scissile bond in full-length VWF. We have used differential scanning fluorimetry together with site-directed mutagenesis of residues involved in both the vicinal disulphide bond and the CBS to demonstrate that both of these sites contribute to stability against thermal unfolding of the isolated VWF A2 domain. Moreover, we show that the combination of site mutations can result in increased susceptibility of FL-VWF to proteolysis by ADAMTS13, even in the absence of an agent (such as urea) required to induce unfolding. These studies demonstrate that VWF A2 domain stability provided by its 2 structural elements (vicinal disulphide bond and CBS) is a key protective determinant against FL-VWF cleavage by ADAMTS13. They suggest a 2-step mechanism for VWF A2 domain unfolding.


Asunto(s)
Proteínas ADAM/metabolismo , Proteolisis , Factor de von Willebrand/química , Factor de von Willebrand/metabolismo , Proteína ADAMTS13 , Sitios de Unión , Calcio/metabolismo , Disulfuros/química , Disulfuros/metabolismo , Células HEK293 , Humanos , Enlace de Hidrógeno , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Unión Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Estabilidad Proteica , Estructura Terciaria de Proteína , Factor de von Willebrand/genética
9.
Blood ; 123(18): 2873-81, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24627527

RESUMEN

Bone morphogenetic protein and activin membrane-bound inhibitor (BAMBI) is a transmembrane protein related to the transforming growth factor-ß superfamily, and is highly expressed in platelets and endothelial cells. We previously demonstrated its positive role in thrombus formation using a zebrafish thrombosis model. In the present study, we used Bambi-deficient mice and radiation chimeras to evaluate the function of this receptor in the regulation of both hemostasis and thrombosis. We show that Bambi(-/-) and Bambi(+/-) mice exhibit mildly prolonged bleeding times compared with Bambi(+/+) littermates. In addition, using 2 in vivo thrombosis models in mesenterium or cremaster muscle arterioles, we demonstrate that Bambi-deficient mice form unstable thrombi compared with Bambi(+/+) mice. No defects in thrombin generation in Bambi(-/-) mouse plasma could be detected ex vivo. Moreover, the absence of BAMBI had no effect on platelet counts, platelet activation, aggregation, or platelet procoagulant function. Similar to Bambi(-/-) mice, Bambi(-/-) transplanted with Bambi(+/+) bone marrow formed unstable thrombi in the laser-induced thrombosis model that receded more rapidly than thrombi that formed in Bambi(+/+) mice receiving Bambi(-/-) bone marrow transplants. Taken together, these results provide strong evidence for an important role of endothelium rather than platelet BAMBI as a positive regulator of both thrombus formation and stability.


Asunto(s)
Endotelio Vascular/metabolismo , Hemostasis , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Trombosis/genética , Trombosis/metabolismo , Animales , Tiempo de Sangría , Coagulación Sanguínea/genética , Plaquetas/metabolismo , Modelos Animales de Enfermedad , Femenino , Genotipo , Masculino , Ratones , Ratones Noqueados , Trombosis/etiología
10.
Blood ; 123(25): 3979-87, 2014 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-24740810

RESUMEN

Protein S is a cofactor for tissue factor pathway inhibitor (TFPI), accelerating the inhibition of activated factor X (FXa). TFPI Kunitz domain 3 residue Glu226 is essential for enhancement of TFPI by protein S. To investigate the complementary functional interaction site on protein S, we screened 44 protein S point, composite or domain swap variants spanning the whole protein S molecule for their TFPI cofactor function using a thrombin generation assay. Of these variants, two protein S/growth arrest-specific 6 chimeras, with either the whole sex hormone-binding globulin (SHBG)-like domain (Val243-Ser635; chimera III) or the SHBG laminin G-type 1 subunit (Ser283-Val459; chimera I), respectively, substituted by the corresponding domain in growth arrest-specific 6, were unable to enhance TFPI. The importance of the protein S SHBG-like domain (and its laminin G-type 1 subunit) for binding and enhancement of TFPI was confirmed in FXa inhibition assays and using surface plasmon resonance. In addition, protein S bound to C4b binding protein showed greatly reduced enhancement of TFPI-mediated inhibition of FXa compared with free protein S. We show that binding of TFPI to the protein S SHBG-like domain enables TFPI to interact optimally with FXa on a phospholipid membrane.


Asunto(s)
Lipoproteínas/metabolismo , Proteína S/metabolismo , Sitios de Unión/genética , Western Blotting , Proteína de Unión al Complemento C4b/metabolismo , Factor Xa/metabolismo , Células HEK293 , Humanos , Lipoproteínas/genética , Mutación , Fosfolípidos/metabolismo , Proteína C/metabolismo , Proteína S/genética , Globulina de Unión a Hormona Sexual/metabolismo , Resonancia por Plasmón de Superficie , Trombina/metabolismo , Tromboplastina/metabolismo
11.
Blood ; 121(18): 3543-4, 2013 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-23640994

RESUMEN

In this issue of Blood, Schiele et al report the development of a monoclonal antibody that reverses the anticoagulant effect of the direct thrombin inhibitor dabigatran.


Asunto(s)
Antídotos/química , Antídotos/farmacología , Antitrombinas/antagonistas & inhibidores , Bencimidazoles/antagonistas & inhibidores , beta-Alanina/análogos & derivados , Animales , Dabigatrán , Masculino , beta-Alanina/antagonistas & inhibidores
13.
Blood ; 129(1): 10-11, 2017 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-28057673
14.
Blood ; 120(25): 5059-62, 2012 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-23074276

RESUMEN

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.


Asunto(s)
Lipoproteínas/genética , Lipoproteínas/metabolismo , Mutación Puntual , Proteína S/metabolismo , Factor Xa/metabolismo , Inhibidores del Factor Xa , Humanos , Lipoproteínas/química , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Resonancia por Plasmón de Superficie , Trombina/metabolismo
15.
Blood ; 119(6): 1555-60, 2012 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-22110247

RESUMEN

VWF and ADAMTS13 are major determinants of platelet adhesion after vessel injury. In the present study, we aimed to determine whether VWF or ADAMTS13 plasma antigen levels influence the risks of ischemic stroke (IS) or myocardial infarction (MI) in young women and how these risks are affected by oral contraceptive (OC) use. VWF and ADAMTS13 plasma antigen levels were measured in a frequency-matched case-control study of 1018 young (18-49 years) women including 175 IS patients and 205 MI patients. Increasing levels of VWF and decreasing levels of ADAMTS13 were associated with the risk of IS and MI in a dose-dependent manner. Having both high VWF and low ADAMTS13 resulted in an odds ratio (OR) of 6.9 (95% confidence interval [95% CI], 2.0-23.0) for IS and 11.3 (95% CI, 3.6-35.2) for MI. Use of OCs increased the risk of IS and MI associated with high VWF (OR = 12; 95% CI, 5.5-26.2 and OR = 7.5, 95% CI, 3.6-15.7, respectively) and the risk of IS associated with low ADAMTS13 (OR = 5.8, 95% CI, 2.7-12.4). We conclude that high VWF and low ADAMTS13 plasma levels both increase the risk of IS and MI. The risks associated with high VWF or low ADAMTS13 levels are further increased by the use of OCs.


Asunto(s)
Proteínas ADAM/sangre , Anticonceptivos Orales/efectos adversos , Infarto del Miocardio/sangre , Accidente Cerebrovascular/sangre , Factor de von Willebrand/metabolismo , Proteína ADAMTS13 , Adolescente , Adulto , Isquemia Encefálica/complicaciones , Estudios de Casos y Controles , Femenino , Humanos , Persona de Mediana Edad , Infarto del Miocardio/etiología , Oportunidad Relativa , Medición de Riesgo , Factores de Riesgo , Accidente Cerebrovascular/etiología , Adulto Joven
16.
Proc Natl Acad Sci U S A ; 108(28): 11602-7, 2011 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-21705658

RESUMEN

The platelet-tethering function of von Willebrand factor (VWF) is proteolytically regulated by ADAMTS13 (a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13), which cleaves the Tyr1605-Met1606 (P1-P1') bond in the VWF A2 domain. To date, most of the functional interactions between ADAMTS13 and VWF that have been characterized involve VWF residues that are C terminal to the scissile bond. We now demonstrate that the substrate P3 position in VWF, Leu1603, is a critical determinant of VWF proteolysis. When VWF Leu1603 was substituted with Ser, Ala, Asn, or Lys in a short VWF substrate, VWF115, proteolysis was either greatly reduced or ablated (up to 400-fold reduction in k(cat)/K(m)). As Leu1603 must interact with residues proximate to the Zn(2+) ion coordinated in the active center of ADAMTS13, we sought the corresponding S3 interacting residues. Substitution of 10 candidate residues in the metalloprotease domain of ADAMTS13 identified two spatially separated clusters centered on Leu198 or Val195 (acting with Leu232 and Leu274, or with Leu151, respectively), as possible subsites interacting with VWF. These experimental findings using the short VWF115 substrate were replicated using full-length VWF. It is hypothesized that VWF Leu1603 interacts with ADAMTS13 Leu198/Leu232/Leu274 and that Val195/Leu151 may form part of a S1 subsite. The recognition of VWF Leu1603 by ADAMTS13, in conjunction with previously reported remote exosites C terminal of the cleavage site, suggests a mechanism whereby the VWF P1-P1' scissile bond is brought into position over the active site for cleavage. Together with recently characterized remote exosite interactions, these findings provide a general framework for understanding the ADAMTS family substrate interactions.


Asunto(s)
Proteínas ADAM/metabolismo , Factor de von Willebrand/metabolismo , Proteínas ADAM/química , Proteínas ADAM/genética , Proteína ADAMTS13 , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Sitios de Unión/genética , Dominio Catalítico/genética , Células HEK293 , Humanos , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , Factor de von Willebrand/química , Factor de von Willebrand/genética
18.
Blood ; 118(12): 3212-21, 2011 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-21715306

RESUMEN

von Willebrand factor (VWF) is a large adhesive glycoprotein with established functions in hemostasis. It serves as a carrier for factor VIII and acts as a vascular damage sensor by attracting platelets to sites of vessel injury. VWF size is important for this latter function, with larger multimers being more hemostatically active. Functional imbalance in multimer size can variously cause microvascular thrombosis or bleeding. The regulation of VWF multimeric size and platelet-tethering function is carried out by ADAMTS13, a plasma metalloprotease that is constitutively active. Unusually, protease activity of ADAMTS13 is controlled not by natural inhibitors but by conformational changes in its substrate, which are induced when VWF is subject to elevated rheologic shear forces. This transforms VWF from a globular to an elongated protein. This conformational transformation unfolds the VWF A2 domain and reveals cryptic exosites as well as the scissile bond. To enable VWF proteolysis, ADAMTS13 makes multiple interactions that bring the protease to the substrate and position it to engage with the cleavage site as this becomes exposed by shear. This article reviews recent literature on the interaction between these 2 multidomain proteins and provides a summary model to explain proteolytic regulation of VWF by ADAMTS13.


Asunto(s)
Proteínas ADAM , Coagulación Sanguínea/fisiología , Trombosis/metabolismo , Factor de von Willebrand , Proteínas ADAM/metabolismo , Proteína ADAMTS13 , Animales , Sitios de Unión , Plaquetas/citología , Plaquetas/metabolismo , Activación Enzimática , Hemorragia/metabolismo , Hemorragia/patología , Humanos , Cinética , Ratones , Ratones Transgénicos , Modelos Moleculares , Unión Proteica , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Reología , Especificidad por Sustrato , Trombosis/patología , Factor de von Willebrand/química , Factor de von Willebrand/metabolismo
19.
Blood ; 117(24): 6685-93, 2011 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-21508412

RESUMEN

Protein S has an important anticoagulant function by acting as a cofactor for activated protein C (APC). We recently reported that the EGF1 domain residue Asp95 is critical for APC cofactor function. In the present study, we examined whether additional interaction sites within the Gla domain of protein S might contribute to its APC cofactor function. We examined 4 residues, composing the previously reported "Face1" (N33S/P35T/E36A/Y39V) variant, as single point substitutions. Of these protein S variants, protein S E36A was found to be almost completely inactive using calibrated automated thrombography. In factor Va inactivation assays, protein S E36A had 89% reduced cofactor activity compared with wild-type protein S and was almost completely inactive in factor VIIIa inactivation; phospholipid binding was, however, normal. Glu36 lies outside the ω-loop that mediates Ca(2+)-dependent phospholipid binding. Using mass spectrometry, it was nevertheless confirmed that Glu36 is γ-carboxylated. Our finding that Gla36 is important for APC cofactor function, but not for phospholipid binding, defines a novel function (other than Ca(2+) coordination/phospholipid binding) for a Gla residue in vitamin K-dependent proteins. It also suggests that residues within the Gla and EGF1 domains of protein S act cooperatively for its APC cofactor function.


Asunto(s)
Ácido 1-Carboxiglutámico/fisiología , Proteína C/metabolismo , Proteína S/metabolismo , Proteína S/fisiología , Ácido 1-Carboxiglutámico/química , Secuencia de Aminoácidos , Sustitución de Aminoácidos/fisiología , Sitios de Unión/genética , Dominio Catalítico/genética , Células Cultivadas , Factor VIIIa/metabolismo , Factor Va/metabolismo , Humanos , Modelos Moleculares , Proteínas Mutantes/metabolismo , Proteínas Mutantes/fisiología , Unión Proteica/genética , Unión Proteica/fisiología , Proteína C/agonistas , Proteína C/fisiología , Dominios y Motivos de Interacción de Proteínas/genética , Dominios y Motivos de Interacción de Proteínas/fisiología , Proteína S/química , Proteína S/genética , Estructura Terciaria de Proteína , Relación Estructura-Actividad
20.
Blood ; 116(16): 3064-72, 2010 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-20647566

RESUMEN

ADAMTS13 modulates von Willebrand factor (VWF) platelet-tethering function by proteolysis of the Tyr1605-Met1606 bond in the VWF A2 domain. To examine the role of the metalloprotease domain of ADAMTS13 in scissile bond specificity, we identified 3 variable regions (VR1, -2, and -3) in the ADAMTS family metalloprotease domain that flank the active site, which might be important for specificity. Eight composite sequence swaps (to residues in ADAMTS1 or ADAMTS2) and 18 single-point mutants were generated in these VRs and expressed. Swapping VR1 (E184-R193) of ADAMTS13 with that of ADAMTS1 or ADAMTS2 abolished/severely impaired ADAMTS13 function. Kinetic analysis of VR1 point mutants using VWF115 as a short substrate revealed reduced proteolytic function (k(cat)/K(m) reduced by 2- to 10-fold) as a result of D187A, R190A, and R193A substitutions. Analysis of VR2 (F216-V220) revealed a minor importance of this region. Mutants of VR3 (G236-A261) proteolysed wild-type VWF115 normally. However, using either short or full-length VWF substrates containing the P1' M1606A mutation, we identified residues within VR3 (D252-P256) that influence P1' amino acid specificity, we hypothesize, by shaping the S1' pocket. It is concluded that 2 subsites, D187-R193 and D252-P256, in the metalloprotease domain play an important role in cleavage efficiency and site specificity.


Asunto(s)
Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Mutación , Factor de von Willebrand/metabolismo , Proteínas ADAM/química , Proteína ADAMTS13 , Secuencia de Aminoácidos , Línea Celular , Expresión Génica , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Factor de von Willebrand/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA