Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 561
Filter
1.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Article in English | MEDLINE | ID: mdl-33441484

ABSTRACT

Humans express seven heparan sulfate (HS) 3-O-sulfotransferases that differ in substrate specificity and tissue expression. Although genetic studies have indicated that 3-O-sulfated HS modulates many biological processes, ligand requirements for proteins engaging with HS modified by 3-O-sulfate (3-OS) have been difficult to determine. In particular, the context in which the 3-OS group needs to be presented for binding is largely unknown. We describe herein a modular synthetic approach that can provide structurally diverse HS oligosaccharides with and without 3-OS. The methodology was employed to prepare 27 hexasaccharides that were printed as a glycan microarray to examine ligand requirements of a wide range of HS-binding proteins. The binding selectivity of antithrombin-III (AT-III) compared well with anti-Factor Xa activity supporting robustness of the array technology. Many of the other examined HS-binding proteins required an IdoA2S-GlcNS3S6S sequon for binding but exhibited variable dependence for the 2-OS and 6-OS moieties, and a GlcA or IdoA2S residue neighboring the central GlcNS3S. The HS oligosaccharides were also examined as inhibitors of cell entry by herpes simplex virus type 1, which, surprisingly, showed a lack of dependence of 3-OS, indicating that, instead of glycoprotein D (gD), they competitively bind to gB and gC. The compounds were also used to examine substrate specificities of heparin lyases, which are enzymes used for depolymerization of HS/heparin for sequence determination and production of therapeutic heparins. It was found that cleavage by lyase II is influenced by 3-OS, while digestion by lyase I is only affected by 2-OS. Lyase III exhibited sensitivity to both 3-OS and 2-OS.


Subject(s)
Epithelial Cells/metabolism , Heparin Lyase/metabolism , Heparitin Sulfate/metabolism , Herpesvirus 1, Human/metabolism , Sulfates/metabolism , Sulfotransferases/metabolism , Acetylglucosamine/chemistry , Acetylglucosamine/metabolism , Antithrombin III/chemistry , Antithrombin III/genetics , Antithrombin III/metabolism , Binding Sites , Binding, Competitive , Carbohydrate Sequence , Cell Line , Cornea/cytology , Cornea/metabolism , Epithelial Cells/pathology , Epithelial Cells/virology , Factor Xa/chemistry , Factor Xa/genetics , Factor Xa/metabolism , Factor Xa Inhibitors/chemistry , Factor Xa Inhibitors/metabolism , Gene Expression , Glucuronic Acid/chemistry , Glucuronic Acid/metabolism , Heparin Lyase/chemistry , Heparin Lyase/genetics , Heparitin Sulfate/chemistry , Herpesvirus 1, Human/growth & development , Host-Pathogen Interactions/genetics , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Microarray Analysis , Protein Binding , Proteolysis , Small Molecule Libraries , Substrate Specificity , Sulfates/chemistry , Sulfotransferases/chemistry , Sulfotransferases/genetics , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
2.
Molecules ; 28(19)2023 Oct 02.
Article in English | MEDLINE | ID: mdl-37836752

ABSTRACT

Thromboembolic disorders, arising from abnormal coagulation, pose a significant risk to human life in the modern world. The FDA has recently approved several anticoagulant drugs targeting factor Xa (FXa) to manage these disorders. However, these drugs have potential side effects, leading to bleeding complications in patients. To mitigate these risks, coagulation factor IXa (FIXa) has emerged as a promising target due to its selective regulation of the intrinsic pathway. Due to the high structural and functional similarities of these coagulation factors and their inhibitor binding modes, designing a selective inhibitor specifically targeting FIXa remains a challenging task. The dynamic behavior of protein-ligand interactions and their impact on selectivity were analyzed using molecular dynamics simulation, considering the availability of potent and selective compounds for both coagulation factors and the co-crystal structures of protein-ligand complexes. Throughout the simulations, we examined ligand movements in the binding site, as well as the contact frequencies and interaction fingerprints, to gain insights into selectivity. Interaction fingerprint (IFP) analysis clearly highlights the crucial role of strong H-bond formation between the ligand and D189 and A190 in the S1 subsite for FIXa selectivity, consistent with our previous study. This dynamic analysis also reveals additional FIXa-specific interactions. Additionally, the absence of polar interactions contributes to the selectivity for FXa, as observed from the dynamic profile of interactions. A contact frequency analysis of the protein-ligand complexes provides further confirmation of the selectivity criteria for FIXa and FXa, as well as criteria for binding and activity. Moreover, a ligand movement analysis reveals key interaction dynamics that highlight the tighter binding of selective ligands to the proteins compared to non-selective and inactive ligands.


Subject(s)
Factor IXa , Factor Xa , Humans , Factor Xa/chemistry , Factor IXa/metabolism , Molecular Dynamics Simulation , Ligands , Blood Coagulation Factors
3.
J Biol Chem ; 296: 100234, 2021.
Article in English | MEDLINE | ID: mdl-33376137

ABSTRACT

Coagulation factor V (FV) plays an anticoagulant role but serves as a procoagulant cofactor in the prothrombinase complex once activated to FVa. At the heart of these opposing effects is the proteolytic removal of its central B-domain, including conserved functional landmarks (basic region, BR; 963-1008 and acidic region 2, AR2; 1493-1537) that enforce the inactive FV procofactor state. Tissue factor pathway inhibitor α (TFPIα) has been associated with FV as well as FV-short, a physiologically relevant isoform with a shortened B-domain missing the BR. However, it is unclear which forms of FV are physiologic ligands for TFPIα. Here, we characterize the binding and regulation of FV and FV-short by TFPIα via its positively charged C-terminus (TFPIα-BR) and examine how bond cleavage in the B-domain influences these interactions. We show that FV-short is constitutively active and functions in prothrombinase like FVa. Unlike FVa, FV-short binds with high affinity (Kd ∼1 nM) to TFPIα-BR, which blocks procoagulant function unless FV-short is cleaved at Arg1545, removing AR2. Importantly, we do not observe FV binding (µM detection limit) to TFPIα. However, cleavage at Arg709 and Arg1018 displaces the FV BR, exposing AR2 and allowing TFPIα to bind via its BR. We conclude that for full-length FV, the detachment of FV BR from AR2 is necessary and sufficient for TFPIα binding and regulation. Our findings pinpoint key forms of FV, including FV-short, that act as physiologic ligands for TFPIα and establish a mechanistic framework for assessing the functional connection between these proteins.


Subject(s)
Factor V/chemistry , Factor Va/chemistry , Lipoproteins/chemistry , Thrombin/genetics , Blood Coagulation/genetics , Factor V/genetics , Factor Va/genetics , Factor Xa/chemistry , Factor Xa/genetics , Humans , Ligands , Lipoproteins/genetics , Protein Binding/genetics , Protein Domains/genetics , Proteolysis/drug effects , Thrombin/chemistry , Thromboplastin/chemistry , Thromboplastin/genetics
4.
Bioorg Chem ; 121: 105676, 2022 04.
Article in English | MEDLINE | ID: mdl-35202850

ABSTRACT

A combinatorial method was devised and applied for the design and identification of substrate-analogue inhibitors of therapeutically relevant serine proteases, such as thrombin and factor Xa. We conceptualized imino acid derived diketomorpholines as generally applicable key intermediates prepared through solid-phase synthesis and prone to be cleaved with primary amines in a traceless fashion. The approach led to a compound library whose members were prepared under bioassay-compatible conditions and directly subjected to the in situ evaluation, allowing a fast prediction of hit compounds. Highly active inhibitors for serine proteases of the coagulation cascade have been identified. The most potent dual inhibitor, 16K, has a binding affinity of 23.9 nM to thrombin and 32.8 nM to factor Xa.


Subject(s)
Factor Xa , Thrombin , Factor Xa/chemistry , Factor Xa Inhibitors , Serine Endopeptidases/metabolism , Serine Proteases , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/pharmacology
5.
Blood ; 134(8): 699-708, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31133602

ABSTRACT

Ixolaris is a potent tick salivary anticoagulant that binds coagulation factor Xa (FXa) and zymogen FX, with formation of a quaternary tissue factor (TF)/FVIIa/ FX(a)/Ixolaris inhibitory complex. Ixolaris blocks TF-induced coagulation and PAR2 signaling and prevents thrombosis, tumor growth, and immune activation. We present a high-resolution structure and dynamics of Ixolaris and describe the structural basis for recognition of FX. Ixolaris consists of 2 Kunitz domains (K1 and K2) in which K2 is strikingly dynamic and encompasses several residues involved in FX binding. This indicates that the backbone plasticity of K2 is critical for Ixolaris biological activity. Notably, a nuclear magnetic resonance-derived model reveals a mechanism for an electrostatically guided, high-affinity interaction between Ixolaris and FX heparin-binding (pro)exosite, resulting in an allosteric switch in the catalytic site. This is the first report revealing the structure-function relationship of an anticoagulant targeting a zymogen serving as a scaffold for TF inhibition.


Subject(s)
Factor Xa Inhibitors/chemistry , Factor Xa Inhibitors/pharmacology , Factor Xa/metabolism , Salivary Proteins and Peptides/chemistry , Salivary Proteins and Peptides/pharmacology , Animals , Factor Xa/chemistry , Humans , Molecular Docking Simulation , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Protein Domains , Ticks/chemistry
6.
J Pediatr Hematol Oncol ; 43(7): e946-e950, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-33512867

ABSTRACT

INTRODUCTION: Enoxaparin is commonly used to treat pediatric thrombosis. Several small retrospective studies have suggested that infants and young children require higher enoxaparin doses to achieve therapeutic anti-factor Xa levels compared with adults. MATERIALS AND METHODS: This is a retrospective study of hospitalized children who received enoxaparin for the treatment of thrombosis at a free-standing children's hospital. The primary objective was to ascertain the enoxaparin dose required to achieve an anti-factor Xa level of 0.5 to 1.0 U/mL among 4 age groups in a large cohort of infants and young children between 60 days and 5 years of age. RESULTS: A total of 176 infants and children were evaluated. The majority of patients were less than 1 year of age (n=104). An inverse relationship between enoxaparin dose needed to achieve therapeutic anti-factor Xa levels and patient age was noted, particularly in the first year of life. Patients who were 60 days to less than 7 months at the time of enoxaparin initiation (n=73) required the highest mean dose among the age groups at 1.73 mg/kg subcutaneously every 12 hours (P<0.0001). CONCLUSION: Infants and young children require higher doses of enoxaparin to achieve therapeutic anti-factor Xa levels compared with adults.


Subject(s)
Anticoagulants/administration & dosage , Enoxaparin/administration & dosage , Factor Xa Inhibitors/blood , Heparin, Low-Molecular-Weight/blood , Thrombosis/drug therapy , Child, Preschool , Factor Xa/chemistry , Female , Follow-Up Studies , Humans , Infant , Male , Prognosis , Retrospective Studies , Thrombosis/blood , Thrombosis/pathology
7.
J Biol Chem ; 294(7): 2422-2435, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30578302

ABSTRACT

The proteolytic conversion of factor V to factor Va is central for amplified flux through the blood coagulation cascade. Heterodimeric factor Va is produced by cleavage at three sites in the middle of factor V by thrombin, yielding an N terminus-derived heavy chain and a C terminus-derived light chain. Here, we show that light chain formation resulting from the C-terminal cleavage is the rate-limiting step in the formation of fully cleaved Va. This rate-limiting step also corresponded to and was sufficient for the ability of cleaved factor V to bind Xa and assemble into the prothrombinase complex. Meizothrombin, the proteinase intermediate in thrombin formation, cleaves factor V more slowly than does thrombin, resulting in a pronounced defect in the formation of the light chain. A ∼100-fold reduced rate of meizothrombin-mediated light chain formation by meizothrombin corresponded to equally slow production of active cofactor and an impaired ability to amplify flux through the coagulation cascade initiated in plasma. We show that this defect arises from the occlusion of anion-binding exosite 2 in the catalytic domain by the covalently retained propiece in meizothrombin. Our findings provide structural insights into the prominent role played by exosite 2 in the rate-limiting step of factor V activation. They also bear on how factor V is converted into a cofactor capable of assembling into prothrombinase.


Subject(s)
Enzyme Precursors/chemistry , Factor Va/chemistry , Proteolysis , Thrombin/chemistry , Enzyme Precursors/metabolism , Factor Va/metabolism , Factor Xa/chemistry , Factor Xa/metabolism , Humans , Protein Binding , Protein Domains , Thrombin/metabolism
8.
J Biol Chem ; 294(19): 7644-7657, 2019 05 10.
Article in English | MEDLINE | ID: mdl-30918026

ABSTRACT

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.


Subject(s)
Blood Coagulation , Factor V/chemistry , Factor Xa/chemistry , Prothrombin/chemistry , Serpins/chemistry , Thrombin/chemistry , Amino Acid Substitution , Antibodies/chemistry , Factor V/genetics , Factor V/metabolism , Factor Xa/genetics , Factor Xa/metabolism , Humans , Kinetics , Mutation, Missense , Protein C/chemistry , Protein C/metabolism , Prothrombin/genetics , Prothrombin/metabolism , Serpins/genetics , Serpins/metabolism , Thrombin/genetics , Thrombin/metabolism
9.
Bioorg Med Chem Lett ; 30(2): 126585, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31859158

ABSTRACT

Firstly, a series of Isosteviol derivatives were synthesized and evaluated for FXa inhibitory activity. Among these compounds, the inhibitory activity of compounds 22, 35 and 38 on FXa was better than that of Isosteviol. Secondly, surface plasmon resonance (SPR) assays were performed for selected compounds. Compounds 22, 35, 38 have similar kinetic signatures, and affinity values were at µM level. Thirdly, compounds 22 and 35 displayed moderate-to-high anticoagulation activity and showed similar sensitivity to PT and aPTT. These findings will provide new insight into the exploration of FXa inhibition.


Subject(s)
Anticoagulants/chemical synthesis , Diterpenes, Kaurane/chemistry , Factor Xa Inhibitors/chemical synthesis , Factor Xa/chemistry , Anticoagulants/metabolism , Crystallography, X-Ray , Diterpenes, Kaurane/metabolism , Drug Design , Factor Xa/metabolism , Factor Xa Inhibitors/metabolism , Humans , Kinetics , Molecular Conformation , Partial Thromboplastin Time , Prothrombin Time , Structure-Activity Relationship , Surface Plasmon Resonance
10.
Parasitology ; 147(5): 559-565, 2020 04.
Article in English | MEDLINE | ID: mdl-31992384

ABSTRACT

Dirofilaria immitis is a parasitic nematode that survives in the circulatory system of suitable hosts for many years, causing the most severe thromboembolisms when simultaneous death of adult worms occurs. The two main mechanisms responsible for thrombus formation in mammals are the activation and aggregation of platelets and the generation of fibrin through the coagulation cascade. The aim of this work was to study the anticoagulant potential of excretory/secretory antigens from D. immitis adult worms (DiES) on the coagulation cascade of the host. Anticoagulant and inhibition assays respectively showed that DiES partially alter the coagulation cascade of the host and reduce the activity of the coagulation factor Xa, a key enzyme in the coagulation process. In addition, a D. immitis protein was identified by its similarity to the homologous serpin 6 from Brugia malayi as a possible candidate to form an inhibitory complex with FXa by sodium dodecyl sulfate polyacrylamide gel electrophoresis and mass spectrometry. These results indicate that D. immitis could use the anticoagulant properties of its excretory/secretory antigens to control the formation of blood clots in its immediate intravascular habitat as a survival mechanism.


Subject(s)
Anticoagulants/metabolism , Dirofilaria immitis/metabolism , Factor Xa/metabolism , Animals , Anticoagulants/chemistry , Antigens, Helminth/chemistry , Antigens, Helminth/metabolism , Dirofilariasis/parasitology , Dogs , Factor Xa/chemistry , Host-Parasite Interactions , Serpins/chemistry , Serpins/metabolism , Thromboembolism/parasitology
11.
Molecules ; 25(8)2020 Apr 19.
Article in English | MEDLINE | ID: mdl-32325823

ABSTRACT

Coagulation factor Xa and factor XIa are proven to be convenient and crucial protein targets for treatment for thrombotic disorders and thereby their inhibitors can serve as effective anticoagulant drugs. In the present work, we focused on the structure-activity relationships of derivatives of pyrrolo[3,2,1-ij]quinolin-2(1H)-one and an evaluation of their activity against factor Xa and factor XIa. For this, docking-guided synthesis of nine compounds based on pyrrolo[3,2,1-ij]quinolin-2(1H)-one was carried out. For the synthesis of new hybrid hydropyrrolo[3,2,1-ij]quinolin-2(1H)-one derivatives, we used convenient structural modification of both the tetrahydro- and dihydroquinoline moiety by varying the substituents at the C6,8,9 positions. In vitro testing revealed that four derivatives were able to inhibit both coagulation factors and three compounds were selective factor XIa inhibitors. An IC50 value of 3.68 µM for was found for the best factor Xa inhibitor and 2 µM for the best factor XIa inhibitor.


Subject(s)
Factor XIa/chemistry , Factor Xa Inhibitors/chemistry , Factor Xa/chemistry , Pyrroles/chemistry , Quinolines/chemistry , Anticoagulants , Drug Design , Factor XIa/antagonists & inhibitors , Hydrogen Bonding , Inhibitory Concentration 50 , Molecular Docking Simulation , Pyrroles/chemical synthesis , Quinolines/chemical synthesis , Structure-Activity Relationship
12.
Molecules ; 25(22)2020 Nov 13.
Article in English | MEDLINE | ID: mdl-33202772

ABSTRACT

Crotoxin, from the venom of the South American rattlesnake Crotalus durissus terrificus, is a potent heterodimeric presynaptic ß-neurotoxin that exists in individual snake venom as a mixture of isoforms of a basic phospholipase A2 (PLA2) subunit (CBa2, CBb, CBc, and CBd) and acidic subunit (CA1-4). Specific natural mutations in CB isoforms are implicated in functional differences between crotoxin isoforms. The three-dimensional structure of two individual CB isoforms (CBa2, CBc), and one isoform in a crotoxin (CA2CBb) complex, have been previously reported. This study concerns CBd, which by interaction with various protein targets exhibits many physiological or pharmacological functions. It binds with high affinity to presynaptic receptors showing neurotoxicity, but also interacts with human coagulation factor Xa (hFXa), exhibiting anticoagulant effect, and acts as a positive allosteric modulator and corrector of mutated chloride channel, cystic fibrosis transmembrane conductance regulator (CFTR), implicated in cystic fibrosis. Thus, CBd represents a novel family of agents that have potential in identifying new drug leads related to anticoagulant and anti-cystic fibrosis function. We determined here the X-ray structure of CBd and compare it with the three other natural isoforms of CB. The structural role of specific amino acid variations between CB isoforms are analyzed and the structural framework of CB for interaction with protein targets is described.


Subject(s)
Crotoxin/chemistry , Phospholipases A2/chemistry , Animals , Anticoagulants/chemistry , Binding Sites , Blood Coagulation , Chromatography, Ion Exchange , Computational Biology , Crotalus , Crystallography, X-Ray , Cystic Fibrosis/drug therapy , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Dimerization , Factor Xa/chemistry , Humans , Neurotoxins/chemistry , Protein Domains , Protein Interaction Mapping , Protein Isoforms
13.
Molecules ; 25(3)2020 Jan 23.
Article in English | MEDLINE | ID: mdl-31979319

ABSTRACT

The coagulation cascade is the process of the conversion of soluble fibrinogen to insoluble fibrin that terminates in production of a clot. Factor Xa (FXa) is a serine protease involved in the blood coagulation cascade. Moreover, FXa plays a vital role in the enzymatic sequence which ends with the thrombus production. Thrombosis is a common causal pathology for three widespread cardiovascular syndromes: acute coronary syndrome (ACS), venous thromboembolism (VTE), and strokes. In this research a series of N-propargyltetrahydroquinoline and 1,2,3-triazole derivatives as a potential factor Xa (FXa) inhibitor were designed, synthesized, and evaluated for their FXa inhibitor activity, cytotoxicity activity and coagulation parameters. Rational design for the desired novel molecules was performed through protein-ligand complexes selection and ligand clustering. The microwave-assisted synthetic strategy of selected compounds was carried out by using Ullmann-Goldberg, N-propargylation, Mannich addition, Friedel-Crafts, and 1,3-dipolar cycloaddition type reactions under microwave irradiation. The microwave methodology proved to be an efficient way to obtain all novel compounds in high yields (73-93%). Furthermore, a thermochemical analysis, optimization and reactivity indexes such as electronic chemical potential (µ), chemical hardness (η), and electrophilicity (ω) were performed to understand the relationship between the structure and the energetic behavior of all the series. Then, in vitro analysis showed that compounds 27, 29-31, and 34 exhibited inhibitory activity against FXa and the corresponding half maximal inhibitory concentration (IC50) values were calculated. Next, a cell viability assay in HEK293 and HepG2 cell lines, and coagulation parameters (anti FXa, Prothrombin time (PT), activated Partial Thromboplastin Time (aPTT)) of the most active novel molecules were performed to determine the corresponding cytotoxicity and possible action on clotting pathways. The obtained results suggest that compounds 27 and 29 inhibited FXa targeting through coagulation factors in the intrinsic and extrinsic pathways. However, compound 34 may target coagulation FXa mainly by the extrinsic and common pathway. Interestingly, the most active compounds in relation to the inhibition activity against FXa and coagulation parameters did not show toxicity at the performed coagulation assay concentrations. Finally, docking studies confirmed the preferential binding mode of N-propargyltetrahydroquinoline and 1,2,3-triazole derivatives inside the active site of FXa.


Subject(s)
Blood Coagulation/drug effects , Factor Xa Inhibitors/chemical synthesis , Factor Xa Inhibitors/pharmacology , Factor Xa/chemistry , Quinolines/chemistry , Triazoles/chemistry , Aniline Compounds/chemical synthesis , Aniline Compounds/chemistry , Azides/chemical synthesis , Azides/chemistry , Blood Coagulation Tests , Cell Line, Tumor , Cell Survival/drug effects , Drug Design , Factor Xa/metabolism , Factor Xa Inhibitors/chemistry , Humans , Inhibitory Concentration 50 , Ligands , Microwaves , Molecular Docking Simulation , Quinolines/chemical synthesis , Triazoles/chemical synthesis
14.
Molecules ; 25(11)2020 May 29.
Article in English | MEDLINE | ID: mdl-32485894

ABSTRACT

The coronavirus disease, COVID-19, caused by the novel coronavirus SARS-CoV-2, which first emerged in Wuhan, China and was made known to the World in December 2019 turned into a pandemic causing more than 126,124 deaths worldwide up to April 16th, 2020. It has 79.5% sequence identity with SARS-CoV-1 and the same strategy for host cell invasion through the ACE-2 surface protein. Since the development of novel drugs is a long-lasting process, researchers look for effective substances among drugs already approved or developed for other purposes. The 3D structure of the SARS-CoV-2 main protease was compared with the 3D structures of seven proteases, which are drug targets, and docking analysis to the SARS-CoV-2 protease structure of thirty four approved and on-trial protease inhibitors was performed. Increased 3D structural similarity between the SARS-CoV-2 main protease, the HCV protease and α-thrombin was found. According to docking analysis the most promising results were found for HCV protease, DPP-4, α-thrombin and coagulation Factor Xa known inhibitors, with several of them exhibiting estimated free binding energy lower than -8.00 kcal/mol and better prediction results than reference compounds. Since some of the compounds are well-tolerated drugs, the promising in silico results may warrant further evaluation for viral anticipation. DPP-4 inhibitors with anti-viral action may be more useful for infected patients with diabetes, while anti-coagulant treatment is proposed in severe SARS-CoV-2 induced pneumonia.


Subject(s)
Anticoagulants/chemistry , Antiviral Agents/chemistry , Betacoronavirus/drug effects , Dipeptidyl-Peptidase IV Inhibitors/chemistry , Protease Inhibitors/chemistry , Viral Nonstructural Proteins/antagonists & inhibitors , Amino Acid Sequence , Anticoagulants/pharmacology , Antiviral Agents/pharmacology , Betacoronavirus/chemistry , Betacoronavirus/enzymology , Betacoronavirus/genetics , Binding Sites , COVID-19 , Coronavirus 3C Proteases , Coronavirus Infections/drug therapy , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Dipeptidyl Peptidase 4/chemistry , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Factor Xa/chemistry , Factor Xa/genetics , Factor Xa/metabolism , Hepacivirus/chemistry , Hepacivirus/enzymology , Hepacivirus/genetics , Humans , Molecular Docking Simulation , Pandemics , Pneumonia, Viral/drug therapy , Protease Inhibitors/pharmacology , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , SARS-CoV-2 , Sequence Alignment , Structural Homology, Protein , Substrate Specificity , Thermodynamics , Thrombin/antagonists & inhibitors , Thrombin/chemistry , Thrombin/genetics , Thrombin/metabolism , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
15.
Anal Biochem ; 580: 62-71, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31091429

ABSTRACT

Chromogenic substrates (CS) are synthetic substrates used to monitor the activity of a target enzyme. It has been reported that some CSs display competitive product inhibition with their target enzyme. Thus, in assays where enzyme activity is continuously monitored over long periods of time, the product inhibition may significantly interfere with the reactions being monitored. Despite this knowledge, it is rare for CSs to be directly incorporated into mathematical models that simulate these assays. This devalues the predictive power of the models. In this study, we examined the interactions between a single enzyme, coagulation factor Xa, and its chromogenic substrate. We developed, and experimentally validated, a mathematical model of a chromogenic assay for factor Xa that explicitly included product inhibition from the CS. We employed Bayesian inference, in the form of Markov-Chain Monte Carlo, to estimate the strength of the product inhibition and other sources of uncertainty such as pipetting error and kinetic rate constants. Our model, together with carefully calibrated biochemistry experiments, allowed for full characterization of the strength and impact of product inhibition in the assay. The effect of CS product inhibition in more complex reaction mixtures was further explored using mathematical models.


Subject(s)
Chromogenic Compounds/chemistry , Factor Xa/chemistry , Models, Theoretical
16.
Protein Expr Purif ; 162: 72-82, 2019 10.
Article in English | MEDLINE | ID: mdl-31022450

ABSTRACT

We present a straightforward, versatile method for expressing and purifying ß-amyloid (Aß40) and transmembrane peptides derived from ß-amyloid precursor protein (Aß55). In principle, these methods should be applicable to other types of strongly aggregating peptides. We start with a DNA plasmid encoding a HexaHis tag with a flexible, hydrophilic linker sequence, followed by a cleavage site, and then Aß peptides. The HexaHis tag rather than a protein fusion partner (e.g., GST) obviates the need for a folded protein in affinity purification. Second, we present two cleavage methods, using either Factor Xa or BNPS-Skatole. Although the latter procedure requires subsequent reduction of the product, we describe methods for minimizing side reactions. Because the use of BNPS-Skatole obviates the need for a folded protein in the cleavage reaction, it is compatible with harsh conditions (e.g., inclusion of detergents and denaturants) needed to solubilize the fusion proteins; such conditions tend to inactivate Factor Xa. Finally, we also describe purification strategies for Aß40 and Aß55 using FPLC and/or reverse phase HPLC. Yields of peptide after these BNPS-Skatole cleavage and peptide reduction, though subquantitative, greatly exceed those obtained using Factor Xa cleavage, as the reaction of BNPS-Skatole is insensitive to the presence of detergents and denaturants, and therefore can be used to produce highly aggregative and low solubility peptides such as Aß55. Trp is a low abundance amino acid in proteins generally, and for peptides like Aß55, and other transmembane peptides lacking Trp in relevant positions, this cleavage method remains a useful option.


Subject(s)
Amyloid beta-Peptides/chemistry , Biochemistry/methods , Amino Acid Sequence , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/isolation & purification , Amyloid beta-Peptides/metabolism , Biocatalysis , Chromatography, Affinity , Chromatography, High Pressure Liquid , Factor Xa/chemistry , Protein Folding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Solubility
17.
Chem Pharm Bull (Tokyo) ; 67(5): 426-432, 2019.
Article in English | MEDLINE | ID: mdl-31061367

ABSTRACT

Quantitative structure-activity relationship (QSAR) techniques, especially those that possess three-dimensional attributes, such as the comparative molecular field analysis (CoMFA), are frequently used in modern-day drug design and other related research domains. However, the requirement for accurate alignment of compounds in CoMFA increases the difficulties encountered in its use. This has led to the development of several techniques-such as VolSurf, Grid-independent descriptors (GRIND), and Anchor-GRIND-which do not require such an alignment. We propose a technique to construct the prediction model that uses molecular interaction field grid potentials as inputs to convolutional neural network. The proposed model has been found to demonstrate higher accuracy compared to the conventional descriptor-based QSAR models as well as Anchor-GRIND techniques. In addition, the method is target independent, and is capable of providing useful information regarding the importance of individual atoms constituting the compounds contained in the chemical dataset used in the proposed analysis. In view of these advantages, the proposed technique is expected to find wide applications in future drug-design operations.


Subject(s)
Neural Networks, Computer , Quantitative Structure-Activity Relationship , Algorithms , Deep Learning , Factor Xa/chemistry , Factor Xa/metabolism , Humans , Ligands , Models, Molecular , Protein Binding
18.
Biochemistry ; 57(32): 4880-4890, 2018 08 14.
Article in English | MEDLINE | ID: mdl-29999301

ABSTRACT

Factor Xa (fXa) inhibition by antithrombin (AT) enabled by heparin or heparan sulfate is critical for controlling blood coagulation. AT activation by heparin has been investigated extensively, while interaction of heparin with trapped AT/fXa intermediates has received relatively little attention. We use native electrospray ionization mass spectrometry to study the role of heparin chains of varying length [hexa-, octa-, deca-, and eicosasaccharides (dp6, dp8, dp10, and dp20, respectively)] in AT/fXa complex assembly. Despite being critical promoters of AT/Xa binding, shorter heparin chains are excluded from the final products (trapped intermediates). However, replacement of short heparin segments with dp20 gives rise to a prominent ionic signal of ternary complexes. These species are also observed when the trapped intermediate is initially prepared in the presence of a short oligoheparin (dp6), followed by addition of a longer heparin chain (dp20), indicating that binding of heparin to AT/fXa complexes takes place after the inhibition event. The importance of the heparin chain length for its ability to associate with the trapped intermediate suggests that the binding likely occurs in a bidentate fashion (where two distinct segments of oligoheparin make contacts with the protein components, while the part of the chain separating these two segments is extended into solution to minimize electrostatic repulsion). This model is corroborated by both molecular dynamics simulations with an explicit solvent and ion mobility measurements in the gas phase. The observed post-inhibition binding of heparin to the trapped AT/fXa intermediates hints at the likely role played by heparan sulfate in their catabolism.


Subject(s)
Antithrombins/chemistry , Factor Xa/chemistry , Glycosaminoglycans/chemistry , Blood Coagulation , Chromatography, Gel , Heparin/chemistry , Humans , Mass Spectrometry
19.
J Biol Chem ; 292(40): 16513-16520, 2017 10 06.
Article in English | MEDLINE | ID: mdl-28743742

ABSTRACT

Antithrombin mainly inhibits factor Xa and thrombin. The reactive center loop (RCL) is crucial for its interactions with its protease targets and is fully inserted into the A-sheet after its cleavage, causing translocation of the covalently linked protease to the opposite end of the A-sheet. Antithrombin variants with altered RCL hinge residues behave as substrates rather than inhibitors, resulting in stoichiometries of inhibition greater than one. Other antithrombin residues have been suggested to interfere with RCL insertion or the stability of the antithrombin-protease complex, but available crystal structures or mutagenesis studies have failed to identify such residues. Here, we characterized two mutations, S365L and I207T, present in individuals with type II antithrombin deficiency and identified a new antithrombin functional domain. S365L did not form stable complexes with thrombin or factor Xa, and the I207T/I207A variants inhibited both proteases with elevated stoichiometries of inhibition. Close proximity of Ile-207 and Ser-365 to the inserted RCL suggested that the preferred reaction of these mutants as protease substrates reflects an effect on the rate of the RCL insertion and protease translocation. However, both residues lie within the final docking site for the protease in the antithrombin-protease complex, supporting the idea that the enhanced substrate reactions may result from an increased dissociation of the final complexes. Our findings demonstrate that the distal end of the antithrombin A-sheet is crucial for the last steps of protease inhibition either by affecting the rate of RCL insertion or through critical interactions with proteases at the end of the A-sheet.


Subject(s)
Antithrombin Proteins/chemistry , Blood Coagulation Disorders, Inherited , Factor Xa/chemistry , Molecular Docking Simulation , Thrombin/chemistry , Amino Acid Substitution , Antithrombin Proteins/genetics , Antithrombin Proteins/metabolism , Catalytic Domain , Factor Xa/genetics , Factor Xa/metabolism , Female , Humans , Male , Mutation, Missense , Protein Domains , Protein Structure, Secondary , Thrombin/genetics , Thrombin/metabolism
20.
J Biol Chem ; 292(35): 14625-14635, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28717005

ABSTRACT

Lipid oxidation due to oxidative stress plays an important role in the pathogenesis of inflammatory and thrombotic cardiovascular diseases. Several findings suggest that lipid peroxidation can alter the function of coagulation proteins and contribute to a hypercoagulable state, but the molecular mechanisms are unclear. Here, we report that oxidized phospholipids suppress the anticoagulant function of the serpin, protein Z-dependent protease inhibitor (ZPI), a specific inhibitor of membrane-associated factor Xa (FXa) that requires protein Z (PZ), phospholipid, and calcium as cofactors. We found that this suppression arises from a diminished ability of the oxidized membrane to function as a cofactor to promote ZPI inhibition of membrane-bound FXa, due fully or in part to the susceptibility of the bound ZPI-PZ complex to oxidative inactivation. Surprisingly, free ZPI was also susceptible to inactivation by oxidized membrane vesicles in the absence of calcium. Oxidized vesicles containing both phosphatidylserine and polyunsaturated fatty acids were required to promote inactivation of the ZPI-PZ complex or free ZPI, indicating that binding of the PZ-complexed or free ZPI to peroxide-modified phospholipid vesicles mediates the inactivation. Heparin protected the ZPI-PZ complex and free ZPI from inactivation, suggesting that blocking the heparin-binding site on ZPI interferes with ZPI binding to lipid or to PZ. This was confirmed by direct lipid-binding experiments. Native PAGE indicated that oxidization induced dissociation of the ZPI-PZ complex and increased the negative charge of ZPI. We conclude that compromised ZPI anticoagulant function could contribute to thrombus initiation and growth in oxidative stress-induced cardiovascular diseases.


Subject(s)
Blood Coagulation , Blood Proteins/metabolism , Factor Xa/metabolism , Lipid Bilayers/metabolism , Lipid Peroxidation , Models, Biological , Serpins/metabolism , Anticoagulants/chemistry , Anticoagulants/metabolism , Anticoagulants/pharmacology , Binding Sites , Binding, Competitive , Blood Coagulation/drug effects , Blood Proteins/agonists , Blood Proteins/chemistry , Calcium Signaling , Factor Xa/chemistry , Heparin/chemistry , Heparin/metabolism , Heparin/pharmacology , Humans , Kinetics , Lipid Bilayers/chemistry , Oxidative Stress/drug effects , Phosphatidylcholines/chemistry , Phosphatidylcholines/metabolism , Phosphatidylserines/chemistry , Phosphatidylserines/metabolism , Protein Multimerization/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Serpins/agonists , Serpins/chemistry , Serpins/genetics , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL