Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 320
Filtrar
1.
Int J Biol Macromol ; 266(Pt 2): 131065, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38521329

RESUMEN

Protein C inhibitor (PCI) maintains hemostasis by inhibiting both procoagulant and anticoagulant serine proteases, and plays important roles in coagulation, fibrinolysis, reproduction, and anti-angiogenesis. The reactive site loop of PCI traps and irreversibly inhibits the proteases like APC (activating protein C), thrombin (FIIa) and factor Xa (FXa). Previous studies on antithrombin (ATIII) had identified Tyr253 and Glu255 as functional exosites that interact and aid in the inhibition of factor IXa and FXa. Presence of exosite in PCI is not known, however a sequence comparison with the PCI from different vertebrate species and ATIII identified Glu239 to be absolutely conserved. PCI residues analogous to ATIII exosite residues were mutated to R238A and E239A. Purified variant PCI in the presence of heparin (10 µg/ml) showed a 2-4 fold decrease in the rate of inhibition of the proteases. However, the stoichiometry of inhibition of FIIa, APC, and FXa by native PCI, R238A and E239A variants were found to be close to 1.0, which also indicated the formation of stable complexes based on SDS-PAGE and western blot analysis with thrombin and APC. Our findings revealed the possible presence of an exosite in PCI that influences the protease inhibition rates.


Asunto(s)
Heparina , Inhibidor de Proteína C , Serina Proteasas , Inhibidor de Proteína C/química , Inhibidor de Proteína C/metabolismo , Heparina/química , Heparina/farmacología , Humanos , Serina Proteasas/metabolismo , Serina Proteasas/química , Trombina/metabolismo , Proteína C/metabolismo , Proteína C/química , Factor Xa/metabolismo , Factor Xa/química , Secuencia de Aminoácidos , Activación Enzimática/efectos de los fármacos
2.
Int J Biol Macromol ; 185: 644-653, 2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34217741

RESUMEN

Highly specific graphene-DNA interactions have been at the forefront of graphene-based sensor design for various analytes, including DNA itself. However, in addition to its detection, DNA also needs to be characterized according to its size and concentration in a sample, which is an additional analytical step. Designing a highly sensitive and selective DNA sensing and characterization platform is, thus, of great interest. The present study demonstrates that a bio-derived, naturally fluorescent protein C-phycoerythrin (CPE) - graphene oxide (GO) bio-composite can be used to detect dsDNA in nanomolar quantities efficiently via fluorescent "turn off/on" mechanism. Interaction with GO temporarily quenches CPE fluorescence in a dose-dependent manner. Analytical characterization indicates an indirect charge transfer with a corresponding loss of crystalline GO structure. The fluorescence is regained with the addition of DNA, while other biomolecules do not pose any hinderance in the detection process. The extent of regain is DNA length dependent, and the corresponding calibration curve successfully quantifies the size of an unknown DNA. The incubation time for detection is ~3-5 min. The bio-composite platform also works successfully in a complex biomolecule matrix and cell lysate. However, the presence of serum albumin poses a hinderance in the serum sample. Particle size analysis proves that CPE displacement from GO surface by the incoming DNA is the reason for the 'turn on' response, and that the sensing process is exclusive to dsDNA. This new platform could be an exciting and rapid DNA sensing and characterization tool.


Asunto(s)
ADN/análisis , Grafito/química , Ficoeritrina/química , Proteína C/química , Técnicas Biosensibles , Dispersión Dinámica de Luz , Fluorescencia , Tamaño de la Partícula , Difracción de Rayos X
3.
Biochim Biophys Acta Gen Subj ; 1865(6): 129892, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33722640

RESUMEN

We previously demonstrated that heterozygous Gly197 to Arg mutation in PROC is associated with venous thrombosis due to the mutation abrogating both zymogenic and enzymatic activities of protein C and activated protein C (APC). In this study, we investigated the role of Gly197 on the structure and function of protein C by replacing it with Ala, Lys and Glu in separate constructs. Characterization of protein C mutants indicated their activation by thrombin is improved ~5-20-fold with the order of PC-G197K > PC-G197E > PC-G197A > PC-WT. Interestingly, the cofactor function of thrombomodulin (TM) in promoting the activation of zymogens by thrombin followed the reverse order of PC-WT > PC-G197A > PC-G197E > PC-G197K. The thrombin-generation inhibitory profiles of zymogens in a tissue factor-mediated thrombin generation assay using protein C-deficient plasma with or without supplementation with TM followed the same order of zymogen activation in the purified system. Evaluation of anticoagulant activities of APC derivatives by prothrombinase and aPTT assays revealed a normal activity for APC-G197A but dramatically impaired activity for the other two mutants. In the endothelial cell permeability assay, APC-G197A exhibited normal antiinflammatory activity, but the other two mutants were nearly inactive. These results suggest that Gly197 plays a key role in TM cofactor-dependent protein C activation by thrombin. It facilitates the recognition of protein C by thrombin in the presence of TM but impedes it in the absence of the cofactor. In APC, a small residue at this position is required for the proper folding/reactivity of the active-site pocket of the protease, a hypothesis supported by structural modeling.


Asunto(s)
Antiinflamatorios/farmacología , Anticoagulantes/farmacología , Glicina/genética , Mutación , Proteína C/química , Proteína C/metabolismo , Factor V/metabolismo , Glicina/química , Glicina/metabolismo , Humanos , Mutagénesis Sitio-Dirigida , Proteína C/genética , Conformación Proteica , Relación Estructura-Actividad , Trombina/metabolismo , Trombomodulina/metabolismo
4.
Antioxid Redox Signal ; 34(1): 32-48, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-32705892

RESUMEN

Aims: In hemolysis, which is accompanied by increased levels of labile redox-active heme and is often associated with hemostatic abnormalities, a decreased activity of activated protein C (APC) is routinely detected. APC is a versatile enzyme that exerts its anticoagulant function through inactivation of clotting factors Va and VIIIa. APC has not been demonstrated to be affected by heme as described for other clotting factors and, thus, is a subject of investigation. Results: We report the interaction of heme with APC and its impact on the protein function by employing spectroscopic and physiologically relevant methods. Binding of heme to APC results in inhibition of its amidolytic and anticoagulant activity, increase of the peroxidase-like activity of heme, and protection of human umbilical vein endothelial cells from heme-induced hyperpermeability. To define the sites that are responsible for heme binding, we mapped the surface of APC for potential heme-binding motifs. T285GWGYHSSR293 and W387IHGHIRDK395, both located on the basic exosite, turned out as potential heme-binding sites. Molecular docking employing a homology model of full-length APC indicated Tyr289 and His391 as the Fe(III)-coordinating amino acids. Innovation: The results strongly suggest that hemolysis-derived heme may directly influence the protein C pathway through binding to APC, conceivably explaining the decreased activity of APC under hemolytic conditions. Further, these results extend our understanding of heme as a multifaceted effector molecule within coagulation and may allow for an improved understanding of disease development in hemostasis under hemolytic conditions. Conclusion: Our study identifies APC as a heme-binding protein and provides insights into the functional consequences.


Asunto(s)
Hemo/química , Hemo/metabolismo , Proteína C/química , Proteína C/metabolismo , Sitios de Unión , Coagulación Sanguínea , Hemólisis , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Relación Estructura-Actividad
5.
Int J Lab Hematol ; 43(2): 266-272, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32964666

RESUMEN

INTRODUCTION: Genetic mutations of PROC and PROS1 are well-known risk factors for deep venous thrombosis (DVT) in the Asian population. However, the genetic profile of Vietnamese patients with DVT remains elusive. This study aimed to investigate the spectrum of genetic mutations of these two genes in Vietnamese patients diagnosed with idiopathic DVT. MATERIALS AND METHODS: A total of 50 Vietnamese patients diagnosed with idiopathic DVT were recruited in this study. The entire coding regions of the protein C and protein S genes were amplified and directly sequenced to determine genetic alterations. RESULTS: Four and six genetic mutations were detected in protein C and protein S genes, respectively, in 24 Vietnamese DVT patients. PROC c.565C > T (p.R189W) was the most common mutation found in 13 out of 50 patients, while the mutations of PROS1 comprised three missense and three nonsense variants which diffuse along the gene. CONCLUSIONS: This study shows that mutations of protein C and protein S genes are prevalent in Vietnamese patients diagnosed with idiopathic DVT, and PROC c.565C > T (p.R189W) was the most common genetic alteration.


Asunto(s)
Predisposición Genética a la Enfermedad , Mutación , Proteína C/genética , Proteína S/genética , Trombosis de la Vena/epidemiología , Trombosis de la Vena/genética , Adulto , Alelos , Sustitución de Aminoácidos , Análisis Mutacional de ADN , Femenino , Estudios de Asociación Genética , Pruebas Genéticas , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Modelos Moleculares , Reacción en Cadena de la Polimerasa , Vigilancia de la Población , Pronóstico , Proteína C/química , Conformación Proteica , Proteína S/química , Relación Estructura-Actividad , Trombosis de la Vena/diagnóstico , Vietnam/epidemiología
6.
J Biol Chem ; 295(45): 15236-15244, 2020 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-32855236

RESUMEN

Activated protein C is a trypsin-like protease with anticoagulant and cytoprotective properties that is generated by thrombin from the zymogen precursor protein C in a reaction greatly accelerated by the cofactor thrombomodulin. The molecular details of this activation remain elusive due to the lack of structural information. We now fill this gap by providing information on the overall structural organization of these proteins using single molecule FRET and small angle X-ray scattering. Under physiological conditions, both zymogen and protease adopt a conformation with all domains vertically aligned along an axis 76 Å long and maximal particle size of 120 Å. This conformation is stabilized by binding of Ca2+ to the Gla domain and is affected minimally by interaction with thrombin. Hence, the zymogen protein C likely interacts with the thrombin-thrombomodulin complex through a rigid body association that produces a protease with essentially the same structural architecture. This scenario stands in contrast to an analogous reaction in the coagulation cascade where conversion of the zymogen prothrombin to the protease meizothrombin by the prothrombinase complex is linked to a large conformational transition of the entire protein. The presence of rigid epidermal growth factor domains in protein C as opposed to kringles in prothrombin likely accounts for the different conformational plasticity of the two zymogens. The new structural features reported here for protein C have general relevance to vitamin K-dependent clotting factors containing epidermal growth factor domains, such as factors VII, IX, and X.


Asunto(s)
Precursores Enzimáticos/química , Precursores Enzimáticos/metabolismo , Proteína C/química , Proteína C/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Humanos , Tamaño de la Partícula , Conformación Proteica , Dispersión del Ángulo Pequeño , Difracción de Rayos X
7.
Nat Commun ; 11(1): 2992, 2020 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-32532974

RESUMEN

Activated protein C (APC) is a plasma serine protease with antithrombotic and cytoprotective functions. Based on the hypothesis that specific inhibition of APC's anticoagulant but not its cytoprotective activity can be beneficial for hemophilia therapy, 2 types of inhibitory monoclonal antibodies (mAbs) are tested: A type I active-site binding mAb and a type II mAb binding to an exosite on APC (required for anticoagulant activity) as shown by X-ray crystallography. Both mAbs increase thrombin generation and promote plasma clotting. Type I blocks all APC activities, whereas type II preserves APC's cytoprotective function. In normal monkeys, type I causes many adverse effects including animal death. In contrast, type II is well-tolerated in normal monkeys and shows both acute and prophylactic dose-dependent efficacy in hemophilic monkeys. Our data show that the type II mAb can specifically inhibit APC's anticoagulant function without compromising its cytoprotective function and offers superior therapeutic opportunities for hemophilia.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Hemofilia A/prevención & control , Fragmentos Fab de Inmunoglobulinas/inmunología , Inhibidor de Proteína C/farmacología , Proteína C/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/clasificación , Anticuerpos Monoclonales/inmunología , Tiempo de Sangría , Permeabilidad de la Membrana Celular/efectos de los fármacos , Células Cultivadas , Cristalografía por Rayos X , Hemofilia A/sangre , Hemorragia/prevención & control , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Fragmentos Fab de Inmunoglobulinas/metabolismo , Macaca fascicularis , Masculino , Proteína C/química , Proteína C/inmunología , Proteína C/metabolismo , Inhibidor de Proteína C/sangre , Inhibidor de Proteína C/farmacocinética
8.
J Thromb Haemost ; 17(12): 2056-2068, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31364267

RESUMEN

BACKGROUND: Activated protein C (APC)-mediated inactivation of factor (F)Va is greatly enhanced by protein S. For inactivation to occur, a trimolecular complex among FVa, APC, and protein S must form on the phospholipid membrane. However, direct demonstration of complex formation has proven elusive. OBJECTIVES: To elucidate the nature of the phospholipid-dependent interactions among APC, protein S, and FVa. METHODS: We evaluated binding of active site blocked APC to phospholipid-coated magnetic beads in the presence and absence of protein S and/or FVa. The importance of protein S and FV residues were evaluated functionally. RESULTS: Activated protein C alone bound weakly to phospholipids. Protein S mildly enhanced APC binding to phospholipid surfaces, whereas FVa did not. However, FVa together with protein S enhanced APC binding (>14-fold), demonstrating formation of an APC/protein S/FVa complex. C4b binding protein-bound protein S failed to enhance APC binding, agreeing with its reduced APC cofactor function. Protein S variants (E36A and D95A) with reduced APC cofactor function exhibited essentially normal augmentation of APC binding to phospholipids, but diminished APC/protein S/FVa complex formation, suggesting involvement in interactions dependent upon FVa. Similarly, FVaNara (W1920R), an APC-resistant FV variant, also did not efficiently incorporate into the trimolecular complex as efficiently as wild-type FVa. FVa inactivation assays suggested that the mutation impairs its affinity for phospholipid membranes and with protein S within the complex. CONCLUSIONS: FVa plays a central role in the formation of its inactivation complex. Furthermore, membrane proximal interactions among FVa, APC, and protein S are essential for its cofactor function.


Asunto(s)
Coagulación Sanguínea , Proteínas de Unión al Calcio/metabolismo , Factor Va/metabolismo , Fosfolípidos/metabolismo , Proteína C/metabolismo , Proteína S/metabolismo , Sitios de Unión , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/genética , Activación Enzimática , Factor Va/química , Factor Va/genética , Células HEK293 , Humanos , Modelos Moleculares , Complejos Multiproteicos , Fosfolípidos/química , Unión Proteica , Proteína C/química , Conformación Proteica , Proteína S/química , Proteína S/genética , Relación Estructura-Actividad , Trombina/metabolismo , Tromboplastina/metabolismo
9.
Anal Chem ; 91(10): 6498-6506, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31025853

RESUMEN

Analysis of protein C termini is very important for functional annotations of proteomes, while proteome-wide C termini analysis still poses substantial challenges. Here we described a simple and robust strategy for specific isolation of protein C termini based on LysC digestion and site-selective dimethylation to deplete N-terminal and internal peptides by scavenger materials. The performance of LysC digestion and conditions of site-selective dimethylation and resin coupling were discussed in detail. Then the strategy was successfully applied to the characterization of protein C termini of HeLa cells. A total of 781 protein C termini were identified with a 300 µg digest in our study, among which 38.9% were actually not identifiable using current trypsin digestion-based methods due to their inappropriate peptide length for MS analysis, indicating that our method was highly complementary to the existing methods. The enrichment procedure was rapid and easy to operate and could afford a very good identification efficiency by obtaining the largest C termini data set of the human proteome with the least sample loading. This method was without bias toward physicochemical properties of peptides. Moreover, a peptide-centric database was first introduced to analyze protein C termini, which effectively improved the accuracy and speed of the database search. Therefore, our method can be used to effectively and selectively isolate protein C termini and contributes to the global annotation of C terminomes.


Asunto(s)
Proteína C/química , Serina Endopeptidasas/metabolismo , Cromatografía Líquida de Alta Presión , Regulación de la Expresión Génica , Células HeLa , Humanos , Proteoma/análisis , Proteómica/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Espectrometría de Masas en Tándem
10.
J Biol Chem ; 294(19): 7644-7657, 2019 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-30918026

RESUMEN

Current thought holds that factor Xa (FXa) bound in the prothrombinase complex is resistant to regulation by protein protease inhibitors during prothrombin activation. Here we provide evidence that, contrary to this view, the FXa-specific serpin inhibitor, protein Z-dependent protease inhibitor (ZPI), complexed with its cofactor, protein Z (PZ), functions as a physiologically significant inhibitor of prothrombinase-bound FXa during prothrombin activation. Kinetics studies showed that the rapid rate of inhibition of FXa by the ZPI-PZ complex on procoagulant membrane vesicles (ka(app) ∼107 m-1 s-1) was decreased ∼10-fold when FXa was bound to FVa in prothrombinase and a further ∼3-4-fold when plasma levels of S195A prothrombin were present (ka(app) 2 × 105 m-1 s-1). Nevertheless, the ZPI-PZ complex produced a major inhibition of thrombin generation during prothrombinase-catalyzed activation of prothrombin under physiologically relevant conditions. The importance of ZPI-PZ complex anticoagulant regulation of FXa both before and after incorporation into prothrombinase was supported by thrombin generation assays in plasma. These showed enhanced thrombin generation when the inhibitor was neutralized with a PZ-specific antibody and decreased thrombin generation when exogenous ZPI-PZ complex was added whether prothrombin was activated directly by FXa or through extrinsic or intrinsic pathway activators. Moreover, the PZ antibody enhanced thrombin generation both in the absence and presence of activated protein C (APC) anticoagulant activity. Taken together, these results suggest an important anticoagulant role for the ZPI-PZ complex in regulating both free FXa generated in the initiation phase of coagulation as well as prothrombinase-bound FXa in the propagation phase that complement prothrombinase regulation by APC.


Asunto(s)
Coagulación Sanguínea , Factor V/química , Factor Xa/química , Protrombina/química , Serpinas/química , Trombina/química , Sustitución de Aminoácidos , Anticuerpos/química , Factor V/genética , Factor V/metabolismo , Factor Xa/genética , Factor Xa/metabolismo , Humanos , Cinética , Mutación Missense , Proteína C/química , Proteína C/metabolismo , Protrombina/genética , Protrombina/metabolismo , Serpinas/genética , Serpinas/metabolismo , Trombina/genética , Trombina/metabolismo
11.
Clin Chem Lab Med ; 57(6): 873-882, 2019 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-30485173

RESUMEN

Background Most guidelines and experts recommend against performance of thrombophilia testing in general, and specifically against testing patients on pharmacological anticoagulants, due to substantially increased risk of false positive identification. For example, vitamin K antagonist (VKA) therapy affects protein C (PC) and protein S (PS), as well as some clotting assays (e.g. as used to investigate activated PC resistance [APCR]). Although heparin may also affect clotting assays, most commercial methods contain neutralisers to make them 'insensitive' to therapeutic levels. Direct oral anticoagulants (DOACs) also affect a wide variety of thrombophilia assays, although most reported data has employed artificial in vitro spiked samples. Methods In the current report, data from our facility for the past 2.5 years has been assessed for all 'congenital thrombophilia' related tests, as evaluated against patient anticoagulant status. We processed 10,571 'thrombophilia' related test requests, including antithrombin (AT; n=3470), PC (n=3569), PS (n=3585), APCR (n=2359), factor V Leiden (FVL; n=2659), and prothrombin gene mutation (PGM; n=2103). Results As expected, VKA therapy affected PC and PS, and despite manufacturer claims, also APCR. Most assays, as suggested by manufacturers, were largely resistant to heparin therapy. DOACs' use was associated with falsely low APCR ratios (i.e. FVL-like effect) and somewhat unexpectedly, anti-Xa agents apixaban and rivaroxaban were also associated with lower AT and higher PS values. Conclusions It is concluded that ex-vivo data appears to confirm the potential for both false positive and false negative 'thrombophilia' events in patients on anticoagulant (including DOAC) treatment.


Asunto(s)
Anticoagulantes/análisis , Pruebas de Coagulación Sanguínea/métodos , Trombofilia/tratamiento farmacológico , Resistencia a la Proteína C Activada/diagnóstico , Administración Oral , Anticoagulantes/uso terapéutico , Antitrombinas/química , Factor V/química , Reacciones Falso Negativas , Reacciones Falso Positivas , Humanos , Proteína C/química , Proteína S/química , Trombofilia/diagnóstico , Warfarina/uso terapéutico
12.
Blood ; 132(2): 159-169, 2018 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-29866816

RESUMEN

Protein C is a plasma serine protease zymogen whose active form, activated protein C (APC), exerts potent anticoagulant activity. In addition to its antithrombotic role as a plasma protease, pharmacologic APC is a pleiotropic protease that activates diverse homeostatic cell signaling pathways via multiple receptors on many cells. Engineering of APC by site-directed mutagenesis provided a signaling selective APC mutant with 3 Lys residues replaced by 3 Ala residues, 3K3A-APC, that lacks >90% anticoagulant activity but retains normal cell signaling activities. This 3K3A-APC mutant exerts multiple potent neuroprotective activities, which require the G-protein-coupled receptor, protease activated receptor 1. Potent neuroprotection in murine ischemic stroke models is linked to 3K3A-APC-induced signaling that arises due to APC's cleavage in protease activated receptor 1 at a noncanonical Arg46 site. This cleavage causes biased signaling that provides a major explanation for APC's in vivo mechanism of action for neuroprotective activities. 3K3A-APC appeared to be safe in ischemic stroke patients and reduced bleeding in the brain after tissue plasminogen activator therapy in a recent phase 2 clinical trial. Hence, it merits further clinical testing for its efficacy in ischemic stroke patients. Recent studies using human fetal neural stem and progenitor cells show that 3K3A-APC promotes neurogenesis in vitro as well as in vivo in the murine middle cerebral artery occlusion stroke model. These recent advances should encourage translational research centered on signaling selective APC's for both single-agent therapies and multiagent combination therapies for ischemic stroke and other neuropathologies.


Asunto(s)
Neuronas/metabolismo , Proteína C/metabolismo , Receptor PAR-1/metabolismo , Animales , Biomarcadores , Barrera Hematoencefálica/metabolismo , Terapia Combinada , Susceptibilidad a Enfermedades , Activación Enzimática , Regulación de la Expresión Génica , Humanos , Proteína C/química , Proteína C/genética , Receptor PAR-1/química , Receptor PAR-1/genética , Regeneración , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/terapia
13.
J Biol Chem ; 292(39): 16249-16256, 2017 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-28801460

RESUMEN

Interactions of soluble proteins with the cell membrane are critical within the blood coagulation cascade. Of particular interest are the interactions of γ-carboxyglutamic acid-rich domain-containing clotting proteins with lipids. Variability among conventional analytical methods presents challenges for comparing clotting protein-lipid interactions. Most previous studies have investigated only a single clotting protein and lipid composition and have yielded widely different binding constants. Herein, we demonstrate that a combination of lipid bilayer nanodiscs and a multiplexed silicon photonic analysis technology enables high-throughput probing of many protein-lipid interactions among blood-clotting proteins. This approach allowed direct comparison of the binding constants of prothrombin, factor X, activated factor VII, and activated protein C to seven different binary lipid compositions. In a single experiment, the binding constants of one protein interacting with all lipid compositions were simultaneously determined. A simple surface regeneration then facilitated similar binding measurements for three other coagulation proteins. As expected, our results indicated that all proteins exhibit tighter binding (lower Kd ) as the proportion of anionic lipid increases. Interestingly, at high proportions of phosphatidylserine, the Kd values of all four proteins began to converge. We also found that although koff values for all four proteins followed trends similar to those observed for the Kd values, the variation among the proteins was much lower, indicating that much of the variation came from the kinetic binding (kon) of the proteins. These findings indicate that the combination of silicon photonic microring resonator arrays and nanodiscs enables rapid interrogation of biomolecular binding interactions at model cell membrane interfaces.


Asunto(s)
Factor VIIa/metabolismo , Factor X/metabolismo , Ácidos Fosfatidicos/metabolismo , Fosfatidilcolinas/metabolismo , Fosfatidilserinas/metabolismo , Proteína C/metabolismo , Protrombina/metabolismo , Factor VIIa/química , Factor VIIa/genética , Factor X/química , Ensayos Analíticos de Alto Rendimiento , Humanos , Cinética , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Nanoestructuras/química , Fenómenos Ópticos , Ácidos Fosfatidicos/química , Fosfatidilcolinas/química , Fosfatidilserinas/química , Análisis por Matrices de Proteínas , Proteína C/química , Protrombina/química , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Silicio/química
14.
J Thromb Haemost ; 15(11): 2198-2207, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28834159

RESUMEN

Essentials The basis of cytoprotective protease-activated receptor 1 (PAR1) signaling is not fully understood. Activated protein C chimera (APCFVII-82 ) was used to identify requirements for PAR1 signaling. APCFVII-82 did not initiate PAR1 signaling, but conferred monocyte anti-inflammatory activity. APC-specific light chain residues are required for cytoprotective PAR1 signaling. SUMMARY: Background Activated protein C (APC) cell signaling is largely reliant upon its ability to mediate protease-activated receptor (PAR) 1 proteolysis when bound to the endothelial cell (EC) protein C (PC) receptor (EPCR). Furthermore, EPCR-bound PC modulates PAR1 signaling by thrombin to induce APC-like EC cytoprotection. Objective The molecular determinants of EPCR-dependent cytoprotective PAR1 signaling remain poorly defined. To address this, a PC-factor VII chimera (PCFVII-82 ) possessing FVII N-terminal domains and conserved EPCR binding was characterized. Methods Activated PC-FVII chimera (APCFVII-82 ) anticoagulant activity was measured with calibrated automated thrombography and activated FV degradation assays. APCFVII-82 signaling activity was characterized by the use of reporter assays of PAR1 proteolysis and EC barrier integrity. APCFVII-82 anti-inflammatory activity was assessed according to its inhibition of nuclear factor-κB (NF-κB) activation and cytokine secretion from monocytes. Results PCFVII-82 was activated normally by thrombin on ECs, but was unable to inhibit plasma thrombin generation. Surprisingly, APCFVII-82 did not mediate EPCR-dependent PAR1 proteolysis, confer PAR1-dependent protection of thrombin-induced EC barrier disruption, or limit PAR1-dependent attenuation of interleukin-6 release from lipopolysaccharide (LPS)-stimulated macrophages. Interestingly, EPCR occupation by active site-blocked APCFVII-82 was, like FVII, unable to mimic EC barrier stabilization induced by PC upon PAR1 proteolysis by thrombin. APCFVII-82 did, however, diminish LPS-induced NF-κB activation and tumor necrosis factor-α release from monocytes in an apolipoprotein E receptor 2-dependent manner, with similar efficacy as wild-type APC. Conclusions These findings identify a novel role for APC light chain amino acid residues outside the EPCR-binding site in enabling cytoprotective PAR1 signaling.


Asunto(s)
Células Endoteliales/metabolismo , Factor VII/metabolismo , Inflamación/prevención & control , Macrófagos/metabolismo , Monocitos/metabolismo , Proteína C/metabolismo , Receptor PAR-1/metabolismo , Animales , Sitios de Unión , Coagulación Sanguínea , Permeabilidad Capilar , Receptor de Proteína C Endotelial/metabolismo , Factor VII/química , Factor VII/genética , Células HEK293 , Humanos , Inflamación/metabolismo , Interleucina-6/metabolismo , Proteínas Relacionadas con Receptor de LDL/metabolismo , Ratones , FN-kappa B/metabolismo , Unión Proteica , Proteína C/química , Proteína C/genética , Dominios y Motivos de Interacción de Proteínas , Células RAW 264.7 , Receptor PAR-1/química , Proteínas Recombinantes de Fusión/química , Transducción de Señal , Relación Estructura-Actividad , Trombina/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
15.
J Thromb Haemost ; 15(7): 1241-1250, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28671348

RESUMEN

Factor V (FV) is a regulator of both pro- and anticoagulant pathways. It circulates as a single-chain procofactor, which is activated by thrombin or FXa to FVa that serves as cofactor for FXa in prothrombin activation. The cofactor function of FVa is regulated by activated protein C (APC) and protein S. FV can also function as an anticoagulant APC cofactor in the inhibition of FVIIIa in the membrane-bound tenase complex (FIXa/FVIIIa). In recent years, it has become clear that FV also functions in multiple ways in the tissue factor pathway inhibitor (TFPI) anticoagulant pathway. Of particular importance is a FV splice variant (FV-Short) that serves as a carrier and cofactor to TFPIα in the inhibition of FXa. FV-Short is generated through alternative splicing of exon 13 that encodes the large activation B domain. A highly negatively charged binding site for TFPIα is exposed in the C-terminus of the FV-Short B domain, which binds the positively charged C-terminus of TFPIα, thus keeping TFPIα in circulation. The binding of TFPIα to FV-Short is also instrumental in localizing the inhibitor to the surface of negatively charged phospholipids, where TFPIα inhibits FXa in process that is stimulated by protein S. Plasma FV activation intermediates and partially proteolyzed platelet FV similarly bind TFPIα with high affinity and regulate formation of prothrombinase. The novel insights gained into the interaction between FV isoforms, TFPIα, and protein S have opened a new avenue for research about the mechanisms of coagulation regulation and also for future development of therapeutics aimed at modulating coagulation.


Asunto(s)
Factor V/química , Lipoproteínas/química , Proteína S/química , Empalme Alternativo , Anticoagulantes/química , Sitios de Unión , Coagulación Sanguínea , Pruebas de Coagulación Sanguínea , Exones , Factor VIIIa/química , Humanos , Mutación Puntual , Unión Proteica , Proteína C/química , Dominios Proteicos , Isoformas de Proteínas/química , Trombina/química , Trombofilia/metabolismo , Tromboplastina/metabolismo
16.
Biochemistry ; 56(24): 3119-3128, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28549218

RESUMEN

Thrombin activity, inhibition, and localization are regulated by two exosites that flank the active site. Substrates, cofactors, and inhibitors bind to exosite 1 to promote active site access, whereas exosite 2 interactions hold thrombin on cells, platelets, and proteins. The exosites also serve allosteric roles, whereby ligand binding alters thrombin activity. Previously, we showed that ligands that bind exosite 2 attenuate the exosite 1-mediated interaction of thrombin with fibrin, demonstrating allosteric connection between the exosites. To determine the functional consequences of these inter-exosite interactions, we examined the effect of exosite 2 ligands on thrombin's interaction with thrombomodulin, a key cofactor that binds exosite 1 and redirects thrombin activity to the anticoagulant protein C pathway. Exosite 2-directed ligands, which included the HD22 aptamer, glycoprotein 1bα-derived peptide, and fibrinogen γ'-chain peptide, reduced the level of exosite 1-mediated thrombin binding to the thrombomodulin peptide consisting of the fourth, fifth, and sixth epidermal-like growth factor-like domains, decreasing affinity by >10-fold, and attenuated thrombomodulin-dependent activation of protein C by 60-80%. The ligands had similar effects on thrombin-mediated protein C activation with intact soluble thrombomodulin and with thrombomodulin on the surface of cultured endothelial cells. Their activity was exosite 2-specific because it was attenuated when RA-thrombin, a variant lacking exosite 2, was used in place of thrombin. These results indicate that additional reactions mediated by exosite 1 are amenable to regulation by exosite 2 ligation, providing further evidence of inter-exosite allosteric regulation of thrombin activity.


Asunto(s)
Proteína C/metabolismo , Trombina/metabolismo , Trombomodulina/metabolismo , Activación Enzimática , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ligandos , Modelos Moleculares , Proteína C/química , Resonancia por Plasmón de Superficie , Trombina/química , Trombomodulina/química
17.
Thromb Haemost ; 117(7): 1358-1369, 2017 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-28405673

RESUMEN

Protein C is a vitamin K-dependent serine protease zymogen in plasma which upon activation by thrombin in complex with thrombomodulin (TM) down-regulates the clotting cascade by a feedback loop inhibition mechanism. Activated protein C (APC) exerts its anticoagulant function through protein S-dependent degradation of factors Va and VIIIa. We recently identified a venous thrombosis patient whose plasma level of protein C antigen is normal, but its anticoagulant activity is only 34 % of the normal level. Genetic analysis revealed that the proband and her younger brother carry a novel heterozygous mutation c.346G>A, p.Gly74Ser (G74S) in PROC. Thrombin generation assay indicated that the TM-dependent anticoagulant activity of the proband's plasma has been significantly impaired. We expressed protein C-G74S in mammalian cells and characterised its properties in established coagulation assays. We demonstrate that the protein C variant can be normally activated by the thrombin-TM complex and the resulting APC mutant also exhibits normal amidolytic and proteolytic activities toward both FVa and FVIIIa. However, it was discovered the protein S-dependent catalytic activity of APC variant toward both procoagulant cofactors has been significantly impaired. Protein S concentration-dependence of FVa degradation revealed that the capacity of APC variant to interact with the cofactor has been markedly impaired. The same results were obtained for inactivation of FVa-Leiden suggesting that the protein S-dependent activity of APC variant toward cleavage of Arg-306 site has been adversely affected. These results provide insight into the mechanism through which G74S substitution in APC causes thrombosis in the proband carrying this mutation.


Asunto(s)
Anticoagulantes/sangre , Proteínas Mutantes/sangre , Proteínas Mutantes/genética , Deficiencia de Proteína C/sangre , Deficiencia de Proteína C/genética , Proteína C/genética , Proteína C/metabolismo , Proteína S/metabolismo , Sustitución de Aminoácidos , Femenino , Células HEK293 , Heterocigoto , Humanos , Masculino , Proteínas Mutantes/química , Mutación Missense , Linaje , Proteína C/química , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Trombina/metabolismo , Trombomodulina/sangre , Trombosis de la Vena/sangre , Trombosis de la Vena/genética
18.
Sci Rep ; 7: 44596, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28294177

RESUMEN

In addition to its procoagulant and proinflammatory functions mediated by cleavage of fibrinogen and PAR1, the trypsin-like protease thrombin activates the anticoagulant protein C in a reaction that requires the cofactor thrombomodulin and the endothelial protein C receptor. Once in the circulation, activated protein C functions as an anticoagulant, anti-inflammatory and regenerative factor. Hence, availability of a protein C activator would afford a therapeutic for patients suffering from thrombotic disorders and a diagnostic tool for monitoring the level of protein C in plasma. Here, we present a fusion protein where thrombin and the EGF456 domain of thrombomodulin are connected through a peptide linker. The fusion protein recapitulates the functional and structural properties of the thrombin-thrombomodulin complex, prolongs the clotting time by generating pharmacological quantities of activated protein C and effectively diagnoses protein C deficiency in human plasma. Notably, these functions do not require exogenous thrombomodulin, unlike other anticoagulant thrombin derivatives engineered to date. These features make the fusion protein an innovative step toward the development of protein C activators of clinical and diagnostic relevance.


Asunto(s)
Deficiencia de Proteína C/sangre , Proteína C/aislamiento & purificación , Proteínas Recombinantes de Fusión/genética , Trombomodulina/química , Receptor de Proteína C Endotelial/química , Receptor de Proteína C Endotelial/genética , Humanos , Péptidos/sangre , Péptidos/química , Proteína C/química , Proteína C/genética , Deficiencia de Proteína C/genética , Proteínas Recombinantes de Fusión/química , Trombomodulina/genética
19.
Biochim Biophys Acta Bioenerg ; 1858(4): 325-335, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28188780

RESUMEN

The cold adaptation mechanism of phycobiliproteins, the major photosynthetic pigment-proteins in cyanobacteria and red algae, has rarely been studied. Here we reported the biochemical, structural, and molecular dynamics simulation study of the C-phycocyanin from Arctic cyanobacterial strain Pseudanabaena sp. LW0831. We characterized the phycobilisome components of LW0831 and obtained their gene sequences. Compared to the mesophilic counterpart from Arthrospira platensis (Ar-C-PC), LW0831 C-phycocyanin (Ps-C-PC) has a decreased thermostability (∆Tm of -16°C), one of the typical features of cold-adapted enzymes. To uncover its structural basis, we resolved the crystal structure of Ps-C-PC 1 at 2.04Å. Consistent with the decrease in thermostability, comparative structural analyses revealed decreased intra-trimer and inter-trimer interactions in Ps-C-PC 1, compared to Ar-C-PC. However, comparative molecular dynamics simulations indicated that Ps-C-PC 1 shows similar flexibilities to Ar-C-PC for both the (αß)3 trimer and (αß)6 hexamer. Therefore, the optimization mode is clearly different from cold-adapted enzymes, which usually have increased flexibilities. Detailed analyses demonstrated different optimization modes for the α and ß subunits and it was revealed that hydrophobic interactions are key to this difference, though salt bridges, hydrogen bonds, and surface hydrophobicity are also involved. This study is the first report of the structure of cold-adapted phycobiliproteins and provides insights into the cold-adaptation strategies of non-enzyme proteins.


Asunto(s)
Cianobacterias/química , Fotosíntesis , Ficobiliproteínas/química , Ficocianina/química , Proteína C/química , Frío , Cristalización , Enlace de Hidrógeno , Simulación de Dinámica Molecular , Estabilidad Proteica
20.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 34(1): 10-14, 2017 Feb 10.
Artículo en Chino | MEDLINE | ID: mdl-28186585

RESUMEN

OBJECTIVE: To explore the pathogenesis of protein C deficiency in two pedigrees through mutation detection and model analysis. METHODS: Chromogenic substrate method and enzyme linked immunosorbent assay (ELISA) were used to determine the plasma protein C activity (PC: A) and protein C antigen (PC: Ag) in the two probands and their family members. All of the 9 exons and intron-exon boundaries of the PROC gene were amplified by PCR and analyzed with Sanger sequencing after purification. Corresponding mutate sites of the family members were also amplified and sequenced. The PolyPhen-2 software was used to analyze the perniciousness of the mutations and Clustal X was to analyze the conservatism. The protein model and amino acids interaction of the mutations were analyzed by Swiss-PdbViewer software. RESULTS: The PC: A and PC: Ag of proband 1 was 30% and 35%, while PC:A of his father, mother and aunt were all slightly under the reference range. Two heterozygous missense mutations were found in exons 7 and 5 of the PROC gene, namely c.565 C>T (p.Arg147Trp) and c.383 G>A (p.Gly86Asp). His father and aunt were carriers for c.565 C>T, while his mother had carried c.383 G>A. The PC: A of proband 2 and his son were 50% and 64%, respectively. And they were both positive for p.Arg147Trp. Analysis of PolyPhen-2 indicated that p.Arg147Trp was benign, while p.Gly86Asp was damaging. Clustal X analysis indicated that the p.Arg147Trp was non-conservative, while the p.Gly86Asp was highly conservative. Modeling for the mutant proteins revealed that the simple aromatic ring of Trp147 in p.Arg147Trp destroyed the two hydrogen bonds between Arg147-Lys146 and Arg147-Lys151, and steric hindranted with Arg178. The side chain of Asp86 extended and generated steric clash with Gln90 with the occurrence of p.Gly86Asp. The change of hydrogen bonds and steric effects has altered the spatial configuration of amino acids, which led to unstable mutate proteins and interfered with the secretion. CONCLUSION: Both probands had hereditary protein C deficiencies, for which their parents were all carriers. The heterozygous mutations p.Arg147Trp and p.Gly86Asp were the main cause for PC: A activity decrease. Among these, p.Gly86Asp was discovered for the first time.


Asunto(s)
Mutación , Deficiencia de Proteína C/genética , Proteína C/genética , Secuencia de Bases , Niño , Análisis Mutacional de ADN/métodos , Salud de la Familia , Femenino , Heterocigoto , Humanos , Enlace de Hidrógeno , Masculino , Persona de Mediana Edad , Modelos Moleculares , Linaje , Fenotipo , Proteína C/química , Proteína C/metabolismo , Deficiencia de Proteína C/sangre , Dominios Proteicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...