Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 200
Filtrar
2.
J Thromb Haemost ; 20(3): 589-599, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34927362

RESUMO

BACKGROUND: The regulation of factor X (FX) is critical to maintain the balance between blood coagulation and fluidity. OBJECTIVES: To functionally characterize the role of the FX autolysis loop in the regulation of the zymogen and active form of FX. METHODS: We introduced novel N-linked glycosylations on the surface-exposed loop spanning residues 143-150 (chymotrypsin numbering) of FX. The activity and inhibition of recombinant FX variants was quantified in pure component assays. The in vitro thrombin generation potential of the FX variants was evaluated in FX-depleted plasma. RESULTS: The factor VIIa (FVIIa)-mediated activation and prothrombin activation was reduced, presumably through steric hinderance. Prothrombin activation was, however, recovered in presence of cofactor factor Va (FVa) despite a reduced prothrombinase assembly. The introduced N-glycans exhibited position-specific effects on the interaction with two FXa inhibitors: tissue factor pathway inhibitor (TFPI) and antithrombin (ATIII). Ki for the inhibition by full-length TFPI of these FXa variants was increased by 7- to 1150-fold, whereas ATIII inhibition in the presence of the heparin-analog Fondaparinux was modestly increased by 2- to 15-fold compared with wild-type. When supplemented in zymogen form, the FX variants exhibited reduced thrombin generation activity relative to wild-type FX, whereas enhanced procoagulant activity was measured for activated FXa variants. CONCLUSION: The autolysis loop participates in all aspects of FX regulation. In plasma-based assays, a modest decrease in FX activation rate appeared to knock down the procoagulant response even when down regulation of FXa activity by inhibitors was reduced.


Assuntos
Fator X , Fator Va/química , Fator X/química , Fator Xa/metabolismo , Humanos , Protrombina/química , Tromboplastina/genética , Tromboplastina/metabolismo
3.
J Thromb Haemost ; 20(3): 600-604, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34894064

RESUMO

BACKGROUND: Cellular trauma or activation exposes phosphatidylserine (PS) and the substantially more abundant phospholipid, phosphatidylethanolamine (PE), on the outer layer of the plasma membrane, thereby allowing binding of many blood clotting proteins. We previously proposed the Anything But Choline (ABC) hypothesis to explain how PS and PE synergize to support binding of clotting proteins with gamma-carboxyglutamate (Gla)-rich domains, which posited that each Gla domain binds to a limited number of PS molecules and multiple PE molecules. However, the minimal number of PS molecules required to stably bind a Gla-domain-containing blood clotting protein in the presence of excess PE was unknown. OBJECTIVE: To test the ABC hypothesis for factor X by determining the threshold binding requirement of PS molecules under conditions of PS-PE synergy. METHODS: We used surface plasmon resonance to investigate the stoichiometry of factor X binding to nanoscale membrane bilayers (Nanodiscs) of varying phospholipid composition. RESULTS AND CONCLUSIONS: We quantified 1.05 ± 0.2 PS molecules per bound factor X molecule in Nanodiscs containing a mixture of 10% PS, 60% PE, and 30% phosphatidylcholine. Hence, there appears to be one truly PS-specific binding site per Gla domain, while the remaining membrane binding interactions can be satisfied by PE.


Assuntos
Fator X , Fosfatidilserinas , Sítios de Ligação , Membrana Celular/metabolismo , Fator X/química , Humanos , Fosfatidilserinas/química , Fosfolipídeos/química , Ligação Proteica
4.
Toxicol Lett ; 340: 77-88, 2021 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-33412251

RESUMO

Species within the viperid genus Macrovipera are some of the most dangerous snakes in the Eurasian region, injecting copious amounts of potent venom. Despite their medical importance, the pathophysiological actions of their venoms have been neglected. Particularly poorly known are the coagulotoxic effects and thus the underlying mechanisms of lethal coagulopathy. In order to fill this knowledge gap, we ascertained the effects of venom upon human plasma for Macrovipera lebetina cernovi, M. l. lebetina, M. l. obtusa, M. l. turanica, and M. schweizeri using diverse coagulation analysing protocols. All five were extremely potent in their ability to promote clotting but varied in their relative activation of Factor X, being equipotent in this study to the venom of the better studied, and lethal, species Daboia russelii. The Insoserp European viper antivenom was shown to be highly effective against all the Macrovipera venoms, but performed poorly against the D. russelii venom. Reciprocally, while Daboia antivenoms performed well against D. russelii venom, they failed against Macrovipera venom. Thus despite the two genera sharing a venom phenotype (Factor X activation) driven by the same toxin type (P-IIId snake venom metalloproteases), the surface biochemistries of the toxins differed significantly enough to impede antivenom cross- neutralization. The differences in venom biochemistry were reflected in coagulation co-factor dependence. While both genera were absolutely dependent upon calcium for the activation of Factor X, dependence upon phospholipid varied. The Macrovipera venoms had low levels of dependence upon phospholipid while the Daboia venom was three times more dependent upon phospholipid for the activation of Factor X. This suggests that the sites on the molecular surface responsible for phospholipid dependence, are the same differential sites that prevent inter-genera antivenom cross- neutralization. Due to cold-chain requirements, antivenoms may not be stocked in rural settings where the need is at the greatest. Thus we tested the efficacy of enzyme inhibitor Prinomastat as a field-deployable treatment to stabilise patients while being transported to antivenom stocks, and showed that it was extremely effective in blocking the Factor X activating pathophysiological actions. Marimastat however was less effective. These results thus not only shed light on the coagulopathic mechanisms of Macrovipera venoms, but also provide data critical for evidence-based design of snakebite management strategies.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Venenos de Víboras/toxicidade , Viperidae/fisiologia , Animais , Antivenenos/farmacologia , Fator X/química , Fator X/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Inibidores de Metaloproteinases de Matriz/farmacologia , Compostos Orgânicos/farmacologia , Fosfolipídeos/química , Especificidade da Espécie
5.
J Thromb Haemost ; 18(9): 2202-2204, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32526092

RESUMO

COVID-19 can be associated with coagulopathy (CAC, COVID-19-associated coagulopathy) with a high prothrombotic risk based on an intense inflammatory response to viral infection leading to immunothrombosis through different procoagulant pathways. Emerging evidence suggests that the use of heparin in these patients could be associated with lower mortality. Emicizumab is a bispecific humanized monoclonal antibody that bridges activated factor IX and factor X, thereby restoring the function of missing factor VIIIa in hemophilia A. The use of emicizumab has been associated with thrombotic events in patients who also received high cumulative amounts of activated prothrombin complex concentrates. Although this risk is extremely low, there is a lack of evidence on whether CAC increases the thrombotic risk in patients on emicizumab prophylaxis. We present the case of a patient with severe hemophilia A in prophylaxis treatment with emicizumab; due to the potential thrombotic risk we decided to administer low molecular weight heparin as prophylaxis treatment without any thrombotic or bleeding complications.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , COVID-19/complicações , Hemofilia A/complicações , Hemofilia A/tratamento farmacológico , Tromboembolia Venosa/prevenção & controle , Anticoagulantes/uso terapêutico , Coagulação Sanguínea , Coagulantes , Fator IXa/química , Fator X/química , Seguimentos , Infecções por HIV/complicações , Hepatite C/complicações , Humanos , Inflamação , Linfoma não Hodgkin/complicações , Masculino , Pessoa de Meia-Idade , Trombose , Tromboembolia Venosa/tratamento farmacológico
6.
Haemophilia ; 25(4): 685-692, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30994257

RESUMO

INTRODUCTION: Inherited deficiencies in the coagulation pathway provide diversified models to investigate the molecular bases of perinatal lethality associated with null-like variants. Differently from X-linked haemophilias, homozygous/doubly heterozygous null variants in the rare autosomally inherited deficiency of factor X (FX) might be incompatible with perinatal survival. AIM: To provide experimental evidence about the null/close-to-null FX function. METHODS: The residual secreted (ELISA) and functional (thrombin generation assays) protein levels associated with the novel nonsense (c.1382G>A; p.Trp461Ter) and missense (c.752T>C; p.Leu251Pro) variants, found in the proposita with life-threatening symptoms at birth, were characterized through recombinant (r)FX expression. RESULTS: The rFX-461Ter showed very low secretion and undetectable function. Expression and function of the predicted readthrough-deriving missense variants (rFX-461Tyr, rFX-461Gln) were also severely impaired. These unfavourable features, due to nucleotide and protein sequence constraints, precluded functional readthrough over the 461 stop codon. Differently, the poorly secreted rFX-251Pro variant displayed residual function that was characterized by anti-TFPI aptamer-based amplification or selective inhibition of activated FX function by fondaparinux in plasma and found to be reduced by approximately three orders of magnitude. Similarly to the rFX-251Pro, a group of catalytic domain missense variants cause poorly secreted molecules with modest function in FX-deficient patients with life-threatening symptoms. CONCLUSIONS: Our data, contributing to the knowledge of the very severe FX deficiency forms, support life-saving requirement of trace FX function, clearly exemplified by the dysfunctional but not completely inactive rFX-251Pro variant that, albeit with severely reduced function, is compatible with a residual activity ensuring minimal haemostasis and permitting perinatal survival.


Assuntos
Domínio Catalítico/genética , Fator X/genética , Fator X/metabolismo , Hemorragias Intracranianas/genética , Mutação de Sentido Incorreto , Sequência de Aminoácidos , Fator X/química , Regulação da Expressão Gênica , Células HEK293 , Humanos , Recém-Nascido , Hemorragias Intracranianas/metabolismo , Hemorragias Intracranianas/prevenção & controle , Fenótipo
7.
Thromb Haemost ; 119(6): 871-881, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30919383

RESUMO

The Cys22-Cys27 disulfide bond of factor X (FX) protease domain is not conserved among coagulation factors and its contribution to the physiological haemostasis and implication in the pathogenesis of haemostatic and thrombotic disorders remain to be elucidated. Mutation p.Cys27Ser was identified in a pedigree of congenital FX deficiency and fluorescence labelling study of transiently transfected HEK293 cells showed accumulation of FX p.Cys27Ser within cell, indicating incompetent secretion partially responsible for the FX deficiency. The clotting activity of FX p.Cys27Ser was decreased to about 90% of wild-type, while amidolytic and pro-thrombinase activities (kcat/Km) determined with recombinant FXa mutant were 1.33- and 4.77-fold lower. Molecular dynamic simulations revealed no major change in global structure between FXa p.Cys27Ser and wild-type FXa; however, without the Cys22-Cys27 disulfide bond, the insertion of newly formed N terminal of catalytic domain after the activation cleavage is hindered, perturbing the conformation transition from zymogen to enzyme. The crystal structure of FXa shows that this disulfide bond is solvent accessible, indicating that its stability might be subject to the oxidation/reduction balance. As demonstrated with FX p.Cys27Ser here, Cys22-Cys27 disulfide bond may modulate FX clotting activity, with reduced FX pertaining less pro-coagulant activity.


Assuntos
Deficiência do Fator X/genética , Fator X/metabolismo , Mutação/genética , Coagulação Sanguínea , Cristalização , Cisteína/genética , Dissulfetos/química , Ativação Enzimática/genética , Fator X/química , Fator X/genética , Células HEK293 , Humanos , Simulação de Dinâmica Molecular , Conformação Proteica , Domínios Proteicos/genética , Proteólise , Relação Estrutura-Atividade
8.
Thromb Res ; 173: 4-11, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30453126

RESUMO

Fusion with human serum albumin (HSA), which represents a well-established technique to extend half-life of therapeutic proteins, commonly exploits intervening peptide linkers as key components. Here, we explored the human coagulation factor X (FX) carboxyl-terminal region, previously demonstrated by us to be dispensable for secretion and coagulant activity, as a natural linker for fusion purposes. To test our hypothesis, we compared direct FX-HSA fusion with the designed FX-HSA fusion proteins mimicking the recombinant activated factor VII (rFVIIa)-HSA or factor IX (FIX)-HSA chimeras, both strongly dependent from artificial linkers. Three constructs were produced by direct tandem fusion (FX-HSA) and through flexible (glycine/serine; FX-GS-HSA, mimicking rFVIIa-HSA) or cleavable (incorporating the FX activation site; FX-CL-HSA, mimicking FIX-HSA) linkers. The FX-HSA was efficiently secreted and displayed prolonged plasma persistence in mice. All chimeras possessed remarkable pro-coagulant activity, comparable to FX for FX-HSA (88.7 ±â€¯6.0%) and FX-CL-HSA (98.0 ±â€¯16.4%) or reduced for FX-GS-HSA (55.8 ±â€¯5.4%). Upon incubation with activators, FX-HSA and FX-CL-HSA displayed a correct activation profile while the FX-GS-HSA activation was slightly defective. In fluorogenic-based assays, FX-HSA showed normal activity over time and a specific amidolytic activity (1.0 ±â€¯0.12) comparable to that of FX. Overall, the FX-HSA features indicate that the FX carboxyl-terminal region represents an intrinsic sequence allowing direct tandem fusion. Our results provide the first experimental evidence for i) a coagulation factor fusion protein with biological properties independent from artificial linkers, ii) the suitability of FX carboxyl-terminal region as a natural linker for fusion purposes.


Assuntos
Fator X/química , Fator X/farmacologia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/farmacologia , Albumina Sérica Humana/química , Albumina Sérica Humana/farmacologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Coagulação Sanguínea/efeitos dos fármacos , Fator VIIa/farmacologia , Fator X/genética , Fator X/farmacocinética , Expressão Gênica , Vetores Genéticos/genética , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes/farmacologia , Albumina Sérica Humana/genética , Albumina Sérica Humana/farmacocinética
9.
Structure ; 26(9): 1178-1186.e3, 2018 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-30017565

RESUMO

Despite being initially identified in the blood filtrate, LEKTI is a 15-domain Kazal-type inhibitor mostly known in the regulation of skin desquamation. In the current study, screening of serine proteases in blood coagulation cascade showed that LEKTI domain 4 has inhibitory activity toward only FXIa, whereas LEKTI domain 6 inhibits both FXIa and FXaB (bovine FXa). Nuclear magnetic resonance structural and dynamic experiments plus molecular dynamics simulation revealed that LEKTI domain 4 has enhanced backbone flexibility at the reactive-site loop. A model of the LEKTI-protease complex revealed that FXaB has a narrower S4 pocket compared with FXIa and hence prefers only small side-chain residues at the P4 position, such as Ala in LEKTI domain 6. Mutational studies combined with a molecular complex model suggest that both a more flexible reactive-site loop and a bulky residue at the P4 position make LEKTI domain 4 a weaker but highly selective inhibitor of FXIa.


Assuntos
Fator XI/antagonistas & inibidores , Fator X/antagonistas & inibidores , Inibidor de Serinopeptidase do Tipo Kazal 5/química , Inibidor de Serinopeptidase do Tipo Kazal 5/metabolismo , Animais , Sítios de Ligação , Coagulação Sanguínea , Bovinos , Fator X/química , Fator XI/química , Humanos , Modelos Moleculares , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Ressonância Magnética Nuclear Biomolecular , Inibidor de Serinopeptidase do Tipo Kazal 5/genética , Especificidade por Substrato
10.
Thromb Haemost ; 118(5): 830-841, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29614522

RESUMO

The link between factor (F)VIII and FX is essential for optimum activity of the tenase complex. The interactive site(s) in FVIII for FX remains to be completely clarified, however. We investigated the FVIII A2 domain-FX association that was speculated from inhibitory mechanism(s) by an anti-A2 autoantibody. SDS-PAGE demonstrated that the purified inhibitor IgG recognizing residues 373-562 blocked FXa cleavage at Arg372 in FVIII, and surface-plasmon resonance (SPR)-based assays showed that intact A2 subunit directly bound to FX (Kd; 63 nM). The FVIII structure model indicated possible FX-binding site(s) in residues 400-429 in A2. One peptide corresponding to residues 400-409 competitively inhibited both the A2-FX binding and FVIIIa/FIXa-dependent FXa generation. Covalent cross-linking was observed between this peptide and FX following reaction with EDC (1-ethyl-3-(3-dimethylaminopropyl)-carbodiimide) using SDS-PAGE. K408 and S409 were not evident in N-terminal sequence analysis of the cross-linked product, suggesting that two residues participated in cross-link formation. SPR-based assays using recombinant FVIII mutants with one or both residues substituted to alanine demonstrated that K408A and K408A/S409A had approximately fourfold high Kd values of wild-type (WT-)FVIII. FXa cleavages at Arg372 in both mutants were significantly delayed, suggesting a contribution of K408 for FXa cleavage at Arg372. Furthermore, FXa generation assays with these mutants demonstrated that the Km values were 1.4- to 1.7-fold greater, and overall catalytic efficiency (kcat/Km) was 0.49- to 0.89-fold lower than with WT-FVIII, suggesting a significant contribution of K408 for FVIII-FX interaction in tenase assembly. We concluded that the K408 in the A2 domain provided an interactive-site for FX.


Assuntos
Cisteína Endopeptidases/metabolismo , Fator VIII/metabolismo , Fator X/metabolismo , Hemofilia A/sangue , Proteínas de Neoplasias/metabolismo , Sequência de Aminoácidos , Especificidade de Anticorpos , Autoanticorpos/sangue , Autoanticorpos/imunologia , Ligação Competitiva , Domínio Catalítico , Cisteína Endopeptidases/química , Epitopos , Fator VIII/química , Fator VIII/genética , Fator VIII/imunologia , Fator X/química , Fator Xa/química , Fator Xa/metabolismo , Hemofilia A/diagnóstico , Hemofilia A/imunologia , Humanos , Modelos Moleculares , Mutação , Proteínas de Neoplasias/química , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Relação Estrutura-Atividade
11.
Blood ; 130(23): 2463-2468, 2017 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-29042366

RESUMO

During the last decade, the development of improved and novel approaches for the treatment of hemophilia A has expanded tremendously. These approaches include factor VIII (FVIII) with extended half-life (eg, FVIII-Fc and PEGylated FVIII), monoclonal antibodies targeting tissue factor pathway inhibitor, small interfering RNA to reduce antithrombin expression and the bispecific antibody ACE910/emicizumab. Emicizumab is a bispecific antibody recognizing both the enzyme factor IXa and the substrate factor X. By simultaneously binding enzyme and substrate, emicizumab mimics some part of the function exerted by the original cofactor, FVIII, in that it promotes colocalization of the enzyme-substrate complex. However, FVIII and the bispecific antibody are fundamentally different proteins and subject to different modes of regulation. Here, we will provide an overview of the similarities and dissimilarities between FVIII and emicizumab from a biochemical and mechanistical perspective. Such insight might be useful in the clinical decision making for those who apply emicizumab in their practice now or in the future, particularly in view of the thrombotic complications that have been reported when emicizumab is used in combination with FVIII-bypassing agents.


Assuntos
Anticorpos Biespecíficos/metabolismo , Anticorpos Biespecíficos/farmacologia , Anticorpos Monoclonais Humanizados/metabolismo , Anticorpos Monoclonais Humanizados/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Fator IX/metabolismo , Fator X/metabolismo , Animais , Anticorpos Biespecíficos/química , Anticorpos Biespecíficos/uso terapêutico , Anticorpos Monoclonais Humanizados/química , Anticorpos Monoclonais Humanizados/uso terapêutico , Descoberta de Drogas , Ativação Enzimática , Fator IX/química , Fator VIII/química , Fator VIII/metabolismo , Fator VIII/farmacologia , Fator VIII/uso terapêutico , Fator X/química , Fator XIIIa/metabolismo , Hemofilia A/sangue , Hemofilia A/tratamento farmacológico , Humanos , Complexos Multiproteicos/metabolismo , Ligação Proteica , Especificidade por Substrato
12.
J Thromb Haemost ; 15(10): 2005-2016, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28782177

RESUMO

Essentials Membrane-binding GLA domains of coagulation factors are essential for proper clot formation. Factor X (FX) is specific to phosphatidylserine (PS) lipids through unknown atomic-level interactions. Molecular dynamics simulations were used to develop the first membrane-bound model of FX-GLA. PS binding modes of FX-GLA were described, and potential PS-specific binding sites identified. SUMMARY: Background Factor X (FX) binds to cell membranes in a highly phospholipid-dependent manner and, in complex with tissue factor and factor VIIa (FVIIa), initiates the clotting cascade. Experimental information concerning the membrane-bound structure of FX with atomic resolution has remained elusive because of the fluid nature of cellular membranes. FX is known to bind preferentially to phosphatidylserine (PS). Objectives To develop the first membrane-bound model of the FX-GLA domain to PS at atomic level, and to identify PS-specific binding sites of the FX-GLA domain. Methods Molecular dynamics (MD) simulations were performed to develop an atomic-level model for the FX-GLA domain bound to PS bilayers. We utilized a membrane representation with enhanced lipid mobility, termed the highly mobile membrane mimetic (HMMM), permitting spontaneous membrane binding and insertion by FX-GLA in multiple 100-ns simulations. In 14 independent simulations, FX-GLA bound spontaneously to the membrane. The resulting membrane-bound models were converted from HMMM to conventional membrane and simulated for an additional 100 ns. Results The final membrane-bound FX-GLA model allowed for detailed characterization of the orientation, insertion depth and lipid interactions of the domain, providing insight into the molecular basis of its PS specificity. All binding simulations converged to the same configuration despite differing initial orientations. Conclusions Analysis of interactions between residues in FX-GLA and lipid-charged groups allowed for potential PS-specific binding sites to be identified. This new structural and dynamic information provides an additional step towards a full understanding of the role of atomic-level lipid-protein interactions in regulating the critical and complex clotting cascade.


Assuntos
Membrana Celular/metabolismo , Fator X/metabolismo , Fosfatidilserinas/metabolismo , Ácido 1-Carboxiglutâmico/metabolismo , Animais , Sítios de Ligação , Bovinos , Fator X/química , Cinética , Simulação de Acoplamento Molecular , Fosfatidilserinas/química , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Relação Estrutura-Atividade
13.
J Biol Chem ; 292(39): 16249-16256, 2017 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-28801460

RESUMO

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.


Assuntos
Fator VIIa/metabolismo , Fator X/metabolismo , Ácidos Fosfatídicos/metabolismo , Fosfatidilcolinas/metabolismo , Fosfatidilserinas/metabolismo , Proteína C/metabolismo , Protrombina/metabolismo , Fator VIIa/química , Fator VIIa/genética , Fator X/química , Ensaios de Triagem em Larga Escala , Humanos , Cinética , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Nanoestruturas/química , Fenômenos Ópticos , Ácidos Fosfatídicos/química , Fosfatidilcolinas/química , Fosfatidilserinas/química , Análise Serial de Proteínas , Proteína C/química , Protrombina/química , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Silício/química
14.
Molecules ; 22(4)2017 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-28441767

RESUMO

Enzymes involved in the coagulation process have received great attention as potential targets for the development of oral anti-coagulants. Among these enzymes, coagulation factor Xa (FXa) has remained the center of attention in the last decade. In this study, 16 ginsenosides and two sapogenins were isolated, identified and quantified. To determine the inhibitory potential on FXa, the chromogenic substrates method was used. The assay suggested that compounds 5, 13 and 18 were mainly responsible for the anti-coagulant effect. Furthermore, these three compounds also possessed high thrombin selectivity in the thrombin inhibition assay. Furthermore, Glide XP from Schrödinger was employed for molecular docking to clarify the interaction between the bioactive compounds and FXa. Therefore, the chemical and biological results indicate that compounds 5 (ginsenoside Rg2), 13 (ginsenoside Rg3) and 18 (protopanaxtriol, PPT) are potential natural inhibitors against FXa.


Assuntos
Fator X/química , Inibidores do Fator Xa/química , Ginsenosídeos/química , Panax/química , Extratos Vegetais/química , Sítios de Ligação , Cromatografia Líquida de Alta Pressão , Avaliação Pré-Clínica de Medicamentos , Fator X/antagonistas & inibidores , Inibidores do Fator Xa/isolamento & purificação , Inibidores do Fator Xa/farmacologia , Ginsenosídeos/isolamento & purificação , Ginsenosídeos/farmacologia , Cinética , Simulação de Acoplamento Molecular , Tempo de Tromboplastina Parcial , Extratos Vegetais/isolamento & purificação , Extratos Vegetais/farmacologia , Ligação Proteica , Proteólise , Trombina/antagonistas & inibidores , Trombina/química
15.
Chemphyschem ; 18(8): 898-905, 2017 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-28133881

RESUMO

Computational methods play a key role in modern drug design in the pharmaceutical industry but are mostly based on force fields, which are limited in accuracy when describing non-classical binding effects, proton transfer, or metal coordination. Here, we propose a general fully quantum mechanical (QM) scheme for the computation of protein-ligand affinities. It works on a single protein cutout (of about 1000 atoms) and evaluates all contributions (interaction energy, solvation, thermostatistical) to absolute binding free energy on the highest feasible QM level. The methodology is tested on two different protein targets: activated serine protease factor X (FXa) and tyrosine-protein kinase 2 (TYK2). We demonstrate that the geometry of the model systems can be efficiently energy-minimized by using general purpose graphics processing units, resulting in structures that are close to the co-crystallized protein-ligand structures. Our best calculations at a hybrid DFT level (PBEh-3c composite method) for the FXa ligand set result in an overall mean absolute deviation as low as 2.1 kcal mol-1 . Though very encouraging, an analysis of outliers indicates that the structure optimization level, conformational sampling, and solvation treatment require further improvement.


Assuntos
Fator X/química , Teoria Quântica , Serina Endopeptidases/química , TYK2 Quinase/química , Sítios de Ligação , Fator X/metabolismo , Humanos , Ligantes , Serina Endopeptidases/metabolismo , TYK2 Quinase/metabolismo
16.
J Thromb Haemost ; 15(1): 131-139, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27774726

RESUMO

Essentials Fiix-prothrombin time (PT) monitoring of warfarin measuring factor (F) II and X, is effective. Plasma obtained during warfarin induction and stable phase in Fiix-trial was assayed. Fiix-PT stabilized anticoagulation earlier than monitoring with traditional PT-INR. FVII had little effect on thrombin generation that was mainly determined by FII and FX. SUMMARY: Background The prothrombin time (PT) is equally prolonged by reduction of each of the vitamin K-dependent (VKD) factors (F) II, VII and X. The Fiix-PT is only affected by FII and FX, the main contributors to thrombin generation (TG). Objective To test the hypothesis that variability in warfarin anticoagulation is reduced early during monitoring with the normalized PT-ratio calculated from Fiix-PT (Fiix-International Normalized Ratio [INR]) compared with traditional PT-INR monitoring. Also, that because of its insensitivity to FVII, Fiix-PT more accurately reflects TG when Fiix-INR and PT-INR are discrepant. Methods Samples from Fiix-trial participants monitored with either Fiix-PT or PT were used. VKD coagulation factors and TG were measured in samples from 40 patients during stable anticoagulation and in serial samples obtained from 26 patients during warfarin induction. TG was assessed in relation to selective reduction in single VKD factors. Results During Fiix-warfarin induction full anticoagulation measured as FII or FX activity was achieved at a similar rate to that with PT-warfarin but subsequently stabilized better. Fiix-INR but not PT-INR mirrored total TG during initiation. During induction, FII (R2 = 0.66) and FX (R2 = 0.52) correlated better with TG and with a steeper slope than did FIX (R2 = 0.37) and in particular FVII (R2 = 0.21). In vitro, FII and FX were the main determinants of TG at concentrations observed during VKA anticoagulation, whereas FVII and FIX had little influence. Conclusions Fiix-PT monitoring reduces anticoagulation variability, suggesting that monitoring FVII has a limited role during VKA management. TG is better reflected by Fiix-PT.


Assuntos
Anticoagulantes/uso terapêutico , Fator X/química , Protrombina/química , Varfarina/uso terapêutico , Idoso , Coagulação Sanguínea/efeitos dos fármacos , Fatores de Coagulação Sanguínea/uso terapêutico , Testes de Coagulação Sanguínea/métodos , Método Duplo-Cego , Monitoramento de Medicamentos , Feminino , Hemostáticos/uso terapêutico , Humanos , Coeficiente Internacional Normatizado , Masculino , Pessoa de Meia-Idade , Tempo de Protrombina , Trombina/química , Vitamina K/química
17.
Fish Shellfish Immunol ; 55: 149-58, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27235370

RESUMO

A transcriptome wide analysis of the constructed cDNA library of snakehead murrel Channa striatus revealed a full length cDNA sequence of coagulation factor X. Sequence analysis of C. striatus coagulation factor X (CsFX) showed that the cDNA contained 1232 base pairs (bp) comprising 1209 bp open reading frame (ORF). The ORF region encodes 424 amino acids with a molecular mass of 59 kDa. The polypeptide contains γ-carboxyglutamic acid (GLA) rich domain and two epidermal growth factor (EGF) like domains including EGF-CA domain and serine proteases trypsin signature profile. CsFX exhibited the maximum similarity with fish species such as Stegastes partitus (78%), Poecilia formosa (76%) and Cynoglossus semilaevis (74%). Phylogenetically, CsFX is clustered together with the fish group belonging to Actinopterygii. Secondary structure of factor X includes alpha helix 28.54%, extended strand 20.75%, beta turn 7.78% and random coil 42.92%. A predicted 3D model of CsFX revealed a short α-helix and a Ca(2+) (Gla domain) binding site in the coil. Four disulfide bridges were found in serine protease trypsin profile. Obviously, the highest gene expression (P < 0.05) was noticed in blood. Further, the changes in expression of CsFX was observed after inducing with bacterial (Aeromonas hydrophila) and fungal (Aphanomyces invadans) infections and other synthetic immune stimulants. Variation in blood clotting time (CT), prothrombin time (PT) and activated prothromboplastin time (APTT) was analyzed and compared between healthy and bacterial infected fishes. During infection, PT and APTT showed a declined clotting time due to the raised level of thrombocytes.


Assuntos
Fator X/genética , Doenças dos Peixes/genética , Proteínas de Peixes/genética , Regulação da Expressão Gênica , Infecções por Bactérias Gram-Negativas/veterinária , Perciformes , Aeromonas hydrophila/fisiologia , Sequência de Aminoácidos , Animais , Aphanomyces/fisiologia , Sequência de Bases , Biologia Computacional , DNA Complementar/genética , DNA Complementar/metabolismo , Fator X/química , Fator X/metabolismo , Doenças dos Peixes/imunologia , Doenças dos Peixes/microbiologia , Proteínas de Peixes/química , Proteínas de Peixes/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Infecções por Bactérias Gram-Negativas/genética , Infecções por Bactérias Gram-Negativas/imunologia , Infecções por Bactérias Gram-Negativas/microbiologia , Lipopolissacarídeos/farmacologia , Filogenia , Poli I-C/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Alinhamento de Sequência/veterinária
18.
J Thromb Haemost ; 14(5): 1043-53, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26924677

RESUMO

UNLABELLED: Essentials Simple and fast assaying of different anticoagulants (ACs) is useful in emergent situations. We used highly diluted prothrombin time (dPT) or highly diluted Fiix-PT (dFiix-PT) to assay ACs. Both tests could quantify target specific anticoagulants and warfarin anticoagulation. Improved results were consistently observed with the dFiix-PT compared with the dPT. SUMMARY: Background Assaying anticoagulants is useful in emergency situations or before surgery. Different specific assays are currently needed depending on the anticoagulant. Objectives We hypothesized that levels of warfarin, dabigatran, rivaroxaban, apixaban, and heparins could be measured with use of the diluted prothrombin time (dPT) and diluted Fiix-PT (dFiix-PT), using highly diluted thromboplastin (TP). The latter test is affected only by reduced levels of active factors II and X but corrects test plasma for other deficiencies Methods Increasing TP dilutions were used to identify suitable dilutions to measure dabigatran, rivaroxaban, apixaban, unfractionated heparin (UFH), and enoxaparin. Calibrators containing known amounts of direct oral anticoagulants (DOACs) were used to make standard curves. Citrated plasma samples were obtained from patients taking warfarin or DOACs with known drug concentrations as determined by specific assays. Results The dFiix-PT at a TP dilution of 1:1156 could be used to measure all of the drugs tested at therapeutic concentrations except for fondaparinux. The dPT achieved the same but required two TP dilutions (1:750 and 1:300). The warfarin effect could be assessed by using dFiix-PT at 1:1156 with a PT ratio identical to the international normalized ratio. Six different TPs yielded similar results, but two were less sensitive. Dabigatran, rivaroxaban, and apixaban could be accurately measured in patient samples using both dilute PT assays, but a better correlation was consistently observed between the dFiix-PT and specific assays than with the dPT. Conclusion The dFiix-PT using a single dilution of TP may be suitable to assess the anticoagulant effects of warfarin, dabigatran, rivaroxaban, apixaban, heparin, and enoxaparin.


Assuntos
Testes de Coagulação Sanguínea/métodos , Dabigatrana/sangue , Enoxaparina/sangue , Heparina/sangue , Pirazóis/sangue , Piridonas/sangue , Rivaroxabana/sangue , Varfarina/sangue , Anticoagulantes/química , Doadores de Sangue , Calibragem , Fator X/química , Feminino , Fondaparinux , Humanos , Coeficiente Internacional Normatizado , Masculino , Polissacarídeos/sangue , Protrombina/química , Tempo de Protrombina , Reprodutibilidade dos Testes , Tromboplastina/química
19.
Protein Sci ; 24(10): 1640-8, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26189766

RESUMO

Human coagulation factor X is a central component of the blood coagulation cascade that converts, under its activated form, prothrombin into thrombin. Generation of thrombin is the final step of the clotting cascade that leads to the clot by polymerization of fibrinogen molecules into a fibrin network. Today, research of new by-passing agents of the coagulation may contribute to an increased interest for human factor X, which may, in consequence, lead to the need of a more exhaustive picture of its structural features. Several post-translational modifications of human factor X such as γ-carboxylation/ß-hydroxylation of the N-terminal light chain and N-/O-glycosylation of the activation peptide have been described. But, so far as we know, no comprehensive studies of its post-translational modifications have been reported. In this article we report an exhaustive structural analysis of human factor X by mass spectrometry using successive protein and peptide mapping. Surprisingly, human factor X was found to be mostly O-glucosylated on its light chain at Ser106 position, Ser9 of its activation peptide is phosphorylated at about 30% and its C-terminal heavy chain is fully O-glycosylated at Thr249 by a mucin-type O-glycan (HexNAc-Hex-NeuAc). The knowledge of these post-translational modifications is mandatory for the development of recombinant molecules.


Assuntos
Fator X/química , Fator X/metabolismo , Espectrometria de Massas , Processamento de Proteína Pós-Traducional , Humanos , Peptídeos/química , Peptídeos/metabolismo
20.
Biochemistry ; 54(30): 4665-71, 2015 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-26169722

RESUMO

The blood coagulation cascade is initiated when the cell-surface complex of factor VIIa (FVIIa, a trypsin-like serine protease) and tissue factor (TF, an integral membrane protein) proteolytically activates factor X (FX). Both FVIIa and FX bind to membranes via their γ-carboxyglutamate-rich domains (GLA domains). GLA domains contain seven to nine bound Ca(2+) ions that are critical for their folding and function, and most biochemical studies of blood clotting have employed supraphysiologic Ca(2+) concentrations to ensure saturation of these domains with bound Ca(2+). Recently, it has become clear that, at plasma concentrations of metal ions, Mg(2+) actually occupies two or three of the divalent metal ion-binding sites in GLA domains, and that these bound Mg(2+) ions are required for full function of these clotting proteins. In this study, we investigated how Mg(2+) influences FVIIa enzymatic activity. We found that the presence of TF was required for Mg(2+) to enhance the rate of FX activation by FVIIa, and we used alanine-scanning mutagenesis to identify TF residues important for mediating this response to Mg(2+). Several TF mutations, including those at residues G164, K166, and Y185, blunted the ability of Mg(2+) to enhance the activity of the TF/FVIIa complex. Our results suggest that these TF residues interact with the GLA domain of FX in a Mg(2+)-dependent manner (although effects of Mg(2+) on the FVIIa GLA domain cannot be ruled out). Notably, these TF residues are located within or immediately adjacent to the putative substrate-binding exosite of TF.


Assuntos
Fator VIIa/química , Magnésio/química , Complexos Multiproteicos/química , Tromboplastina/química , Substituição de Aminoácidos , Cálcio/química , Cálcio/metabolismo , Fator VIIa/genética , Fator VIIa/metabolismo , Fator X/química , Fator X/genética , Fator X/metabolismo , Humanos , Magnésio/metabolismo , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Mutação de Sentido Incorreto , Estrutura Terciária de Proteína , Tromboplastina/genética , Tromboplastina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...