Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 137
Filter
1.
Plant Cell Physiol ; 63(12): 1980-1993, 2023 Jan 30.
Article in English | MEDLINE | ID: mdl-34977939

ABSTRACT

Plant viruses cause systemic diseases that severely impair plant growth and development. While the accumulation of viruses in the root system has long been established, little is known as to how viruses affect root architecture. Here, we examined how the emerging tobamovirus, tomato brown rugose fruit virus (ToBRFV), alters root development in tomato. We found that ToBRFV and tobacco mosaic virus both invaded root systems during the first week of infection. ToBRFV infection of tomato plants resulted in a significant decrease in root biomass and elongation and root-to-shoot ratio and a marked suppression of root branching. Mutation in RNA-dependent RNA polymerase 6 increased the susceptibility of tomato plants to ToBRFV, resulting in severe reduction of various root growth parameters including root branching. Viral root symptoms were associated with the accumulation of auxin response factor 10a (SlARF10a) transcript, a homolog of Arabidopsis ARF10, a known suppressor of lateral root development. Interestingly, loss-of-function mutation in SlARF10a moderated the effect of ToBRFV on root branching. In contrast, downregulation of sly-miR160a, which targets SlARF10a, was associated with constitutive suppression root branching independent of viral infection. In addition, overexpression of a microRNA-insensitive mutant of SlARF10a mimicked the effect of ToBRFV on root development, suggesting a specific role for SlARF10a in ToBRFV-mediated suppression of root branching. Taken together, our results provide new insights into the impact of tobamoviruses on root development and the role of ARF10a in the suppression of root branching in tomato.


Subject(s)
Solanum lycopersicum , Tobamovirus , Solanum lycopersicum/genetics , Tobamovirus/genetics , Factor Xa/genetics , Indoleacetic Acids , Mutation , Plant Diseases
2.
Dis Markers ; 2022: 1118195, 2022.
Article in English | MEDLINE | ID: mdl-36438904

ABSTRACT

Background: Mitochondria have been involved in host defense upon viral infections. Factor Xa (FXa), a coagulating factor, may also have influence on mitochondrial functionalities. The aim was to analyze if in human pulmonary microvascular endothelial cells (HPMEC), the SARS-CoV-2 (COVID-19) spike protein subunits, S1 and S2 (S1+S2), could alter mitochondrial metabolism and what is the role of FXA. Methods: HPMEC were incubated with and without recombinants S1+S2 (10 nmol/L each). Results: In control conditions, S1+S2 failed to modify FXa expression. However, in LPS (1 µg/mL)-incubated HPMEC, S1+S2 significantly increased FXa production. LPS tended to reduce mitochondrial membrane potential with respect to control, but in higher and significant degree, it was reduced when S1+S2 were present. LPS did not significantly modify cytochrome c oxidase activity as compared with control. Addition of S1+S2 spike subunits to LPS-incubated HPMEC significantly increased cytochrome c oxidase activity with respect to control. Lactate dehydrogenase activity was also increased by S1+S2 with respect to control and LPS alone. Protein expression level of uncoupled protein-2 (UCP-2) was markedly expressed when S1+S2 were added together to LPS. Rivaroxaban (50 nmol/L), a specific FXa inhibitor, significantly reduced all the above-mentioned alterations induced by S1+S2 including UCP-2 expression. Conclusions: In HPMEC undergoing to preinflammatory condition, COVID-19 S1+S2 spike subunits promoted alterations in mitochondria metabolism suggesting a shift from aerobic towards anaerobic metabolism that was accompanied of high FXa production. Rivaroxaban prevented all the mitochondrial metabolic changes mediated by the present COVID-19 S1 and S2 spike subunits suggesting the involvement of endogenous FXa.


Subject(s)
COVID-19 , Factor Xa Inhibitors , Factor Xa , Mitochondria , Rivaroxaban , Spike Glycoprotein, Coronavirus , Humans , COVID-19/genetics , COVID-19/metabolism , Electron Transport Complex IV/metabolism , Endothelial Cells/metabolism , Factor Xa/genetics , Factor Xa/metabolism , Lipopolysaccharides/pharmacology , Lipopolysaccharides/metabolism , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Protein Subunits/metabolism , Rivaroxaban/metabolism , Rivaroxaban/pharmacology , Rivaroxaban/therapeutic use , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , COVID-19 Drug Treatment , Factor Xa Inhibitors/metabolism , Factor Xa Inhibitors/pharmacology , Factor Xa Inhibitors/therapeutic use , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use
3.
Molecules ; 26(17)2021 Sep 03.
Article in English | MEDLINE | ID: mdl-34500804

ABSTRACT

Blood coagulation is an essential physiological process for hemostasis; however, abnormal coagulation can lead to various potentially fatal disorders, generally known as thromboembolic disorders, which are a major cause of mortality in the modern world. Recently, the FDA has approved several anticoagulant drugs for Factor Xa (FXa) which work via the common pathway of the coagulation cascade. A main side effect of these drugs is the potential risk for bleeding in patients. Coagulation Factor IXa (FIXa) has recently emerged as the strategic target to ease these risks as it selectively regulates the intrinsic pathway. These aforementioned coagulation factors are highly similar in structure, functional architecture, and inhibitor binding mode. Therefore, it remains a challenge to design a selective inhibitor which may affect only FIXa. With the availability of a number of X-ray co-crystal structures of these two coagulation factors as protein-ligand complexes, structural alignment, molecular docking, and pharmacophore modeling were employed to derive the relevant criteria for selective inhibition of FIXa over FXa. In this study, six ligands (three potent, two selective, and one inactive) were selected for FIXa inhibition and six potent ligands (four FDA approved drugs) were considered for FXa. The pharmacophore hypotheses provide the distribution patterns for the principal interactions that take place in the binding site. None of the pharmacophoric patterns of the FXa inhibitors matched with any of the patterns of FIXa inhibitors. Based on pharmacophore analysis, a selectivity of a ligand for FIXa over FXa may be defined quantitatively as a docking score of lower than -8.0 kcal/mol in the FIXa-grids and higher than -7.5 kcal/mol in the FXa-grids.


Subject(s)
Anticoagulants/pharmacology , Factor IXa/antagonists & inhibitors , Factor Xa Inhibitors/pharmacology , Factor Xa/metabolism , Anticoagulants/chemistry , Crystallography, X-Ray , Factor IXa/genetics , Factor IXa/metabolism , Factor Xa/genetics , Factor Xa Inhibitors/chemistry , Humans , Models, Molecular , Molecular Structure
4.
Genome Biol Evol ; 13(1)2021 01 07.
Article in English | MEDLINE | ID: mdl-33527140

ABSTRACT

Bloodfeeding is employed by many parasitic animals and requires specific innovations for efficient feeding. Some of these innovations are molecular features that are related to the inhibition of hemostasis. For example, bloodfeeding insects, bats, and leeches release proteins with anticoagulatory activity through their salivary secretions. The antistasin-like protein family, composed of serine protease inhibitors with one or more antistasin-like domains, is tightly linked to inhibition of hemostasis in leeches. However, this protein family has been recorded also in non-bloodfeeding invertebrates, such as cnidarians, mollusks, polychaetes, and oligochaetes. The present study aims to 1) root the antistasin-like gene tree and delimit the major orthologous groups, 2) identify potential independent origins of salivary proteins secreted by leeches, and 3) identify major changes in domain and/or motif structure within each orthologous group. Five clades containing leech antistasin-like proteins are distinguishable through rigorous phylogenetic analyses based on nine new transcriptomes and a diverse set of comparative data: the trypsin + leukocyte elastase inhibitors clade, the antistasin clade, the therostasin clade, and two additional, unnamed clades. The antistasin-like gene tree supports multiple origins of leech antistasin-like proteins due to the presence of both leech and non-leech sequences in one of the unnamed clades, but a single origin of factor Xa and trypsin + leukocyte elastase inhibitors. This is further supported by three sequence motifs that are exclusive to antistasins, the trypsin + leukocyte elastase inhibitor clade, and the therostasin clade, respectively. We discuss the implications of our findings for the evolution of this diverse family of leech anticoagulants.


Subject(s)
Annelida/genetics , Annelida/metabolism , Evolution, Molecular , Invertebrate Hormones/genetics , Invertebrate Hormones/metabolism , Leeches/genetics , Animals , Anticoagulants/chemistry , Factor Xa/genetics , Hemostasis , Phylogeny , Salivary Proteins and Peptides/genetics , Serine Proteinase Inhibitors/genetics , Transcriptome
5.
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
6.
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
7.
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
8.
Toxicol Lett ; 316: 171-182, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31442586

ABSTRACT

Australian elapid snakes are some of the most venomous snakes in the world and are unique among venomous snakes in having mutated forms of the blood clotting factor X in an activated form (FXa) as a key venom component. In human bite victims, an overdose of this activated clotting enzyme results in the systemic consumption of fibrinogen due to the large amounts of endogenous thrombin generated by the conversion of prothrombin to thrombin by venom FXa. Within Australian elapids, such procoagulant venom is currently known from the tiger snake clade (Hoplocephalus, Notechis, Paroplocephalus, and Tropidechis species), brown/taipan (Oxyuranus and Pseudonaja species) clade, and the red-bellied black snake Pseudechis porphyriacus. We used a STA-R Max coagulation analyser and TEG5000 thromboelastographers to test 47 Australian elapid venoms from 19 genera against human plasma in vitro. In addition to activity being confirmed in the two clades above, FXa-driven potent procoagulant activity was found in four additional genera (Cryptophis, Demansia, Hemiaspis, and Suta). Ontogenetic changes in procoagulant function was also identified as a feature of Suta punctata venom. Phylogenetic analysis of FX sequences confirmed that snake venom FXa toxins evolved only once, that the potency of these toxins against human plasma has increased in a stepwise fashion, and that multiple convergent amplifications of procoagulant activity within Australian elapid snakes have occurred. Cofactor dependence tests revealed all procoagulant venoms in our study, except those of the tiger snake clade, to be highly calcium-dependent, whereas phospholipid dependence was less of a feature but still displayed significant variation between venoms. Antivenom testing using CSL Tiger Snake Antivenom showed broad but differential cross-reactivity against procoagulant venoms, with P. porphyriacus and S. punctata extremely well neutralised but with Cryptophis, Demansia, and Hemiaspis less well-neutralised. The relative variation was not in accordance to genetic relatedness of the species used in antivenom production (Notechis scutatus), which underscores a fundamental principle that the rapid evolution characteristic of venoms results in organismal phylogeny being a poor predictor of antivenom efficacy. Our results have direct and immediate implications for the design of clinical management plans in the event of snakebite by such lesser known Australian elapid snake species that have been revealed in this study to be as potent as the better studied, and proven lethal, species.


Subject(s)
Antivenins/pharmacology , Blood Coagulation/drug effects , Elapid Venoms/antagonists & inhibitors , Elapidae , Factor Xa Inhibitors/pharmacology , Factor Xa/metabolism , Snake Bites/drug therapy , Animals , Cross Reactions , Elapid Venoms/genetics , Elapid Venoms/immunology , Elapid Venoms/metabolism , Elapidae/classification , Elapidae/genetics , Elapidae/immunology , Elapidae/metabolism , Evolution, Molecular , Factor Xa/genetics , Factor Xa/immunology , Fibrinolysis/drug effects , Mutation , Phylogeny , Snake Bites/immunology , Snake Bites/metabolism , Thrombelastography
9.
Molecules ; 24(11)2019 Jun 06.
Article in English | MEDLINE | ID: mdl-31174390

ABSTRACT

Pentamidine is bis-oxybenzamidine-based antiprotozoal drug. The parenteral use of pentamidine appears to affect the processes of blood coagulation and/or fibrinolysis resulting in rare but potentially life-threatening blood clot formation. Pentamidine was also found to cause disseminated intravascular coagulation syndrome. To investigate the potential underlying molecular mechanism(s) of pentamidine's effects on coagulation and fibrinolysis, we studied its effects on clotting times in normal and deficient human plasmas. Using normal plasma, pentamidine isethionate doubled the activated partial thromboplastin time at 27.5 µM, doubled the prothrombin time at 45.7 µM, and weakly doubled the thrombin time at 158.17 µM. Using plasmas deficient of factors VIIa, IXa, XIa, or XIIa, the concentrations to double the activated partial thromboplastin time were similar to that obtained using normal plasma. Pentamidine also inhibited plasmin-mediated clot lysis with half-maximal inhibitory concentration (IC50) value of ~3.6 µM. Chromogenic substrate hydrolysis assays indicated that pentamidine inhibits factor Xa and plasmin with IC50 values of 10.4 µM and 8.4 µM, respectively. Interestingly, it did not significantly inhibit thrombin, factor XIa, factor XIIIa, neutrophil elastase, or chymotrypsin at the highest concentrations tested. Michaelis-Menten kinetics and molecular modeling studies revealed that pentamidine inhibits factor Xa and plasmin in a competitive fashion. Overall, this study provides quantitative mechanistic insights into the in vitro effects of pentamidine isethionate on coagulation and fibrinolysis via the disruption of the proteolytic activity of factor Xa and plasmin.


Subject(s)
Blood Coagulation/drug effects , Fibrinolysis/drug effects , Pentamidine/pharmacology , Thrombosis/drug therapy , Blood Coagulation Tests , Factor VIIa/genetics , Factor XIIa/genetics , Factor XIa/genetics , Factor Xa/genetics , Humans , Partial Thromboplastin Time , Prothrombin Time , Thrombin/chemistry , Thrombin/genetics , Thrombin Time , Thrombosis/blood , Thrombosis/pathology
10.
AAPS J ; 21(3): 52, 2019 04 11.
Article in English | MEDLINE | ID: mdl-30976993

ABSTRACT

This paper presents a systemic investigation of ADA development and ADA impact of a human coagulation factor in nonclinical species during drug development and provides insights into potential implications in human if a similar ADA occurs. FXaI16L-induced ADA response was characterized in monkey, mouse, rat, and dog in different studies, and ADA effects on pharmacokinetic and/or pharmacodynamics of FXaI16L were further examined in ADA-negative and ADA-positive animals. After repeated administrations, FXaI16L elicited a dose and exposure day-dependent ADA response which ranged from no response to a transient or persistent response. Increase in exposure day and increase in dose generally enhanced ADA incidence except for a decrease in ADA incidence was observed in monkeys after repeated high-dose administrations. The observable ADA impact on pharmacokinetics was only found in some ADA+ animals and included decrease in clearance and increase in systemic exposure but no increase in half-life. In addition, no or limited effect on pharmacodynamics by ADA was observed. The earliest ADA response was observed after three exposure days, marked elevation of drug exposure was observed in some animals at log titer > 2.0, and the highest antibody titer excited was about 4 (Log10) in all species. A correlation between ADA induction and accumulative exposure after various repeat treatments in different species was found for FXaI16L. In addition, potential immunogenicity risk and mitigation of ADA in clinics are discussed.


Subject(s)
Factor Xa/immunology , Hemophilia A/drug therapy , Animals , Blood Coagulation/drug effects , Blood Coagulation/immunology , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Factor Xa/administration & dosage , Factor Xa/genetics , Female , Half-Life , Hemophilia A/blood , Hemophilia A/diagnosis , Hemophilia A/immunology , Humans , Macaca fascicularis , Male , Mice , Partial Thromboplastin Time , Prothrombin Time , Rats , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Sequence Homology, Amino Acid , Species Specificity
11.
Expert Opin Biol Ther ; 19(5): 387-397, 2019 05.
Article in English | MEDLINE | ID: mdl-30974977

ABSTRACT

INTRODUCTION: Andexanet alfa is a recombinant modified factor Xa protein that has been developed to reverse factor Xa inhibitors. Since May 2018, the FDA has approved its utilization in patients treated with apixaban and rivaroxaban in case of life-threatening or uncontrolled bleeding. On 28 of February 2019, the Committee for Medicinal Products for Human Use adopted a positive opinion, recommending the granting of a conditional marketing authorization for andexanet alfa in Europe. Area covered: The authors provide an overview of andexanet alfa development and its pharmacokinetic and pharmacodynamic properties. The results of the clinical phase III trial ANNEXA as well as current limitations related to andexanet alfa are also discussed. Expert opinion: Although phase I and II studies have proven that andexanet alfa can be effective in reversing the effect of factor Xa inhibitors, its efficacy in major bleeding patients has only been shown for apixaban and rivaroxaban, without any comparator group. Well-designed studies comparing the efficacy and safety of andexanet alfa to other reversal strategies are required to confirm preliminary data. The benefit of andexanet alfa in specific settings needs to be investigated and its use in clinical practice needs to be facilitated by the implementation of international guidelines.


Subject(s)
Factor Xa Inhibitors/immunology , Factor Xa/therapeutic use , Hemorrhage/drug therapy , Recombinant Proteins/therapeutic use , Clinical Trials as Topic , Factor Xa/genetics , Factor Xa/metabolism , Factor Xa/pharmacokinetics , Half-Life , Humans , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacokinetics
12.
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
13.
Blood Adv ; 2(11): 1300-1314, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29891592

ABSTRACT

Venous thromboembolism (VTE) is a leading cause of morbidity and mortality in elderly people. Increased expression of tumor suppressor protein 53 (p53) has been implicated in vascular senescence. Here, we examined the importance of endothelial p53 for venous thrombosis and whether endothelial senescence and p53 overexpression are involved in the exponential increase of VTE with age. Mice with conditional, endothelial-specific deletion of p53 (End.p53-KO) and their wild-type littermates (End.p53-WT) underwent subtotal inferior vena cava (IVC) ligation to induce venous thrombosis. IVC ligation in aged (12-month-old) End.p53-WT mice resulted in higher rates of thrombus formation and greater mean thrombus size vs adult (12-week-old) End.p53-WT mice, whereas aged End.p53-KO mice were protected from vein thrombosis. Analysis of primary endothelial cells from aged mice or human vein endothelial cells after induction of replicative senescence revealed significantly increased early growth response gene-1 (Egr1) and heparanase expression, and plasma factor Xa levels were elevated in aged End.p53-WT, but not in End.p53-KO mice. Increased endothelial Egr1 and heparanase expression also was observed after doxorubicin-induced p53 overexpression, whereas p53 inhibition using pifithrin-α reduced tissue factor (TF) expression. Importantly, inhibition of heparanase activity using TF pathway inhibitor-2 (TFPI2) peptides prevented the enhanced venous thrombus formation in aged mice and restored it to the thrombotic phenotype of adult mice. Our findings suggest that p53 accumulation and heparanase overexpression in senescent endothelial cells are critically involved in mediating the increased risk of venous thrombosis with age and that heparanase antagonization may be explored as strategy to ameliorate the prothrombotic endothelial phenotype with age.


Subject(s)
Aging/metabolism , Endothelial Cells/metabolism , Tumor Suppressor Protein p53/biosynthesis , Venous Thrombosis/metabolism , Aging/genetics , Aging/pathology , Animals , Early Growth Response Protein 1/biosynthesis , Early Growth Response Protein 1/genetics , Endothelial Cells/pathology , Factor Xa/biosynthesis , Factor Xa/genetics , Glycoproteins/genetics , Glycoproteins/metabolism , Heparin Lyase/genetics , Mice , Mice, Knockout , Tumor Suppressor Protein p53/genetics , Venous Thrombosis/genetics
14.
Nat Biotechnol ; 36(7): 606-613, 2018 08.
Article in English | MEDLINE | ID: mdl-29863725

ABSTRACT

Unfractionated heparin (UFH), the standard anticoagulant for cardiopulmonary bypass (CPB) surgery, carries a risk of post-operative bleeding and is potentially harmful in patients with heparin-induced thrombocytopenia-associated antibodies. To improve the activity of an alternative anticoagulant, the RNA aptamer 11F7t, we solved X-ray crystal structures of the aptamer bound to factor Xa (FXa). The finding that 11F7t did not bind the catalytic site suggested that it could complement small-molecule FXa inhibitors. We demonstrate that combinations of 11F7t and catalytic-site FXa inhibitors enhance anticoagulation in purified reaction mixtures and plasma. Aptamer-drug combinations prevented clot formation as effectively as UFH in human blood circulated in an extracorporeal oxygenator circuit that mimicked CPB, while avoiding side effects of UFH. An antidote could promptly neutralize the anticoagulant effects of both FXa inhibitors. Our results suggest that drugs and aptamers with shared targets can be combined to exert more specific and potent effects than either agent alone.


Subject(s)
Anticoagulants/administration & dosage , Factor Xa Inhibitors/administration & dosage , Factor Xa/chemistry , Postoperative Hemorrhage/drug therapy , Anticoagulants/chemistry , Aptamers, Nucleotide/administration & dosage , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/genetics , Cardiopulmonary Bypass/adverse effects , Crystallography, X-Ray , Drug Combinations , Factor Xa/genetics , Factor Xa Inhibitors/chemistry , Heparin/adverse effects , Humans , Postoperative Hemorrhage/genetics , Postoperative Hemorrhage/pathology , Protein Conformation/drug effects
15.
Nat Commun ; 8(1): 528, 2017 09 13.
Article in English | MEDLINE | ID: mdl-28904343

ABSTRACT

The absence of an adequate reversal strategy to prevent and stop potential life-threatening bleeding complications is a major drawback to the clinical use of the direct oral inhibitors of blood coagulation factor Xa. Here we show that specific modifications of the substrate-binding aromatic S4 subpocket within the factor Xa active site disrupt high-affinity engagement of the direct factor Xa inhibitors. These modifications either entail amino-acid substitution of S4 subsite residues Tyr99 and/or Phe174 (chymotrypsinogen numbering), or extension of the 99-loop that borders the S4 subsite. The latter modifications led to the engineering of a factor Xa variant that is able to support coagulation in human plasma spiked with (supra-)physiological concentrations of direct factor Xa inhibitors. As such, this factor Xa variant has the potential to be employed to bypass the direct factor Xa inhibitor-mediated anticoagulation in patients that require restoration of blood coagulation.A major drawback in the clinical use of the oral anticoagulants that directly inhibit factor Xa in order to prevent blood clot formation is the potential for life threatening bleeding events. Here the authors describe factor Xa variants that are refractory to inhibition by these anticoagulants and could serve as rescue agents in treated patients.


Subject(s)
Factor Xa Inhibitors/chemistry , Factor Xa Inhibitors/pharmacology , Factor Xa/genetics , Factor Xa/metabolism , Blood Coagulation/drug effects , Blood Coagulation Tests , Factor Xa/chemistry , Humans , Kinetics , Protein Engineering
16.
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
17.
J Biol Chem ; 292(22): 9335-9344, 2017 06 02.
Article in English | MEDLINE | ID: mdl-28420729

ABSTRACT

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


Subject(s)
Anticoagulants/metabolism , Blood Coagulation/physiology , Factor V/metabolism , Amino Acid Substitution , Factor V/genetics , Factor Xa/genetics , Factor Xa/metabolism , Humans , Lipoproteins/genetics , Lipoproteins/metabolism , Mutation, Missense , Protein Domains , Protein S/genetics , Protein S/metabolism , Prothrombin/genetics , Prothrombin/metabolism
18.
PLoS One ; 12(3): e0174827, 2017.
Article in English | MEDLINE | ID: mdl-28358853

ABSTRACT

Cleavage activation of the hemagglutinin (HA) protein by host proteases is a crucial step in the infection process of influenza A viruses (IAV). However, IAV exists in eighteen different HA subtypes in nature and their cleavage sites vary considerably. There is uncertainty regarding which specific proteases activate a given HA in the human respiratory tract. Understanding the relationship between different HA subtypes and human-specific proteases will be valuable in assessing the pandemic potential of circulating viruses. Here we utilized fluorogenic peptides mimicking the HA cleavage motif of representative IAV strains causing disease in humans or of zoonotic/pandemic potential and tested them with a range of proteases known to be present in the human respiratory tract. Our results show that peptides from the H1, H2 and H3 subtypes are cleaved efficiently by a wide range of proteases including trypsin, matriptase, human airway tryptase (HAT), kallikrein-related peptidases 5 (KLK5) and 12 (KLK12) and plasmin. Regarding IAVs currently of concern for human adaptation, cleavage site peptides from H10 viruses showed very limited cleavage by respiratory tract proteases. Peptide mimics from H6 viruses showed broader cleavage by respiratory tract proteases, while H5, H7 and H9 subtypes showed variable cleavage; particularly matriptase appeared to be a key protease capable of activating IAVs. We also tested HA substrate specificity of Factor Xa, a protease required for HA cleavage in chicken embryos and relevant for influenza virus production in eggs. Overall our data provide novel tool allowing the assessment of human adaptation of IAV HA subtypes.


Subject(s)
Hemagglutinins/metabolism , Influenza A virus/metabolism , Peptides/metabolism , Factor Xa/genetics , Factor Xa/metabolism , Fibrinolysin/genetics , Fibrinolysin/metabolism , Hemagglutinins/genetics , Humans , Influenza A Virus, H9N2 Subtype/metabolism , Influenza A virus/genetics , Kallikreins/genetics , Kallikreins/metabolism , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Peptides/genetics , Proteolysis , Respiratory System/metabolism , Trypsin/genetics , Trypsin/metabolism
19.
Arterioscler Thromb Vasc Biol ; 36(8): 1525-33, 2016 08.
Article in English | MEDLINE | ID: mdl-27283743

ABSTRACT

OBJECTIVE: The role of hypercoagulability in the pathogenesis of diabetic nephropathy (DN) remains elusive. We recently reported the increased infiltration of macrophages expressing tissue factor in diabetic kidney glomeruli; tissue factor activates coagulation factor X (FX) to FXa, which in turn stimulates protease-activated receptor 2 (PAR2) and causes inflammation. APPROACH AND RESULTS: Here, we demonstrated that diabetes mellitus increased renal FX mRNA, urinary FXa activity, and FX expression in glomerular macrophages. Administration of an oral FXa inhibitor, edoxaban, ameliorated DN with concomitant reductions in the expression of PARs (Par1 and Par2) and of proinflammatory and profibrotic genes. Diabetes mellitus induced PAR2, and lack of Par2 ameliorated DN. FXa or PAR2 agonist increased inflammatory cytokines in endothelial cells and podocytes in vitro. CONCLUSIONS: We conclude that enhanced FXa and PAR2 exacerbate DN and that both are promising targets for preventing DN. Alleviating inflammation is probably more important than inhibiting coagulation per se when treating kidney diseases using anticoagulants.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Diabetic Nephropathies/prevention & control , Factor Xa Inhibitors/pharmacology , Factor Xa/drug effects , Kidney Glomerulus/drug effects , Pyridines/pharmacology , Receptor, PAR-2/metabolism , Thiazoles/pharmacology , Animals , Blood Coagulation/drug effects , Cell Line , Cytokines/genetics , Cytokines/metabolism , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Factor Xa/genetics , Factor Xa/metabolism , Genotype , Humans , Inflammation Mediators/metabolism , Insulin/genetics , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, Knockout , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide Synthase Type III/genetics , Phenotype , Podocytes/drug effects , Podocytes/metabolism , Receptor, PAR-2/antagonists & inhibitors , Receptor, PAR-2/deficiency , Receptor, PAR-2/genetics , Signal Transduction/drug effects , Up-Regulation
20.
Thromb Res ; 141 Suppl 2: S31-3, 2016 May.
Article in English | MEDLINE | ID: mdl-27207419

ABSTRACT

There is a clinical need to develop safe and rapid therapeutic strategies to control bleeding arising from a host of emergent situations. Over the past several years our laboratory has developed novel zymogen-like FXa variants and tested their safety and efficacy using hemophilia as a model system. The variants have a spectrum of properties resulting from an amino acid change at the N-terminus of the heavy chain that alters a critical conformational change. These properties, which include resistance to plasma protease inhibitors, low activity in the absence of FVa, and rescue of low activity upon incorporation in prothrombinase, yield remarkably effective pro-hemostatic agents. The FVa-dependent restoration of activity is a key aspect to their efficacy and also contributes to localizing the variants to the site of vascular injury. While pre-clinical data support their use in the setting of hemophilia, they have the potential to act as rapid pro-hemostatic agents for the treatment of a range of bleeding conditions. This review will discuss the biochemical properties of these FXa zymogen-like variants and their in vivo characterization.


Subject(s)
Enzyme Precursors/therapeutic use , Factor Xa/therapeutic use , Hemophilia A/drug therapy , Hemophilia B/drug therapy , Hemorrhage/drug therapy , Hemostatics/therapeutic use , Animals , Blood Coagulation/drug effects , Enzyme Precursors/chemistry , Enzyme Precursors/genetics , Factor Xa/chemistry , Factor Xa/genetics , Hemophilia A/blood , Hemophilia B/blood , Hemorrhage/blood , Hemostatics/metabolism , Humans , Protein Engineering
SELECTION OF CITATIONS
SEARCH DETAIL