Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 177
Filter
1.
Anal Chem ; 96(40): 16091-16098, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39340422

ABSTRACT

Coagulation factor XIa (FXIa) is associated with a low risk of bleeding and has been identified as an effective and safe target for the development of novel anticoagulant drugs. In this study, we established an ultrasensitive competitive dual-enzyme cascade signal amplification method for the quantitative analysis and screening of FXIa inhibitors. Due to the specific recognition of FXIa's active site by the aptamer AptE40, the AptE40-QDs-EK recognition probe modified with enterokinase (EK) and the aptamer AptE40, was attached to the MNPs-FXIa capture probe. When FXIa inhibitor was present, it competed with AptE40 for binding to FXIa, resulting in the detachment of AptE40-QDs-EK from MNPs-FXIa. After magnetic separation, the enterokinase of AptE40-QDs-EK in the supernatant hydrolyzed N-terminal hexapeptide of trypsinogen, leading to the production of a large amount of trypsin as part of the first-stage signal cascade amplification. Next, trypsin could hydrolyze the hexameric arginine peptide (RRRRRR, R6), leading to the dissociation of RQDs from the R6-RQDs signal probe; this resulted in a dramatic increase in the fluorescence intensity of the supernatant as the second-stage signal cascade was amplified. The feasibility of the method was investigated using the FXIa inhibitor aptamer FELIAP as a positive model drug. Furthermore, the method was applied to screen the FXIa inhibitors in Eupolyphaga sinensis Walker. Two fractions with more active anticoagulated ingredients were successfully identified and validated via the conventional method, and the results were consistent. The established method provides a key technique for the sensitive detection, high-throughput analysis, and screening of the FXIa inhibitors.


Subject(s)
Aptamers, Nucleotide , Factor XIa , Factor XIa/antagonists & inhibitors , Factor XIa/metabolism , Factor XIa/analysis , Humans , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/metabolism , Spectrometry, Fluorescence , Enteropeptidase/metabolism , Enteropeptidase/antagonists & inhibitors , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Fluorescence
2.
J Cardiovasc Pharmacol ; 84(1): 71-80, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38922574

ABSTRACT

ABSTRACT: Clinical practice shows that a critical unmet need in the field of thrombosis prevention is the availability of anticoagulant therapy without bleeding risk. Inhibitors against FXIa or FXIIa have been extensively studied because of their low bleeding risk. However, whether these compounds produce synergistic effects has not yet been explored. In this study, analyses of activated partial thromboplastin time in combination with the FXIa inhibitor PN2KPI and the FXIIa inhibitor Infestin4 at different proportions were performed using the SynergyFinder tool identifying synergistic anticoagulation effects. Both an FeCl 3 -induced carotid artery thrombosis mouse model and a transient occlusion of the middle cerebral artery mouse model showed that the combination of PN2KPI and Infestin4, which are 28.57% and 6.25% of the effective dose, respectively, significantly prevents coagulation, and furthermore, dual inhibition does not cause bleeding risk.


Subject(s)
Anticoagulants , Blood Coagulation , Disease Models, Animal , Drug Synergism , Factor XIIa , Factor XIa , Animals , Factor XIa/antagonists & inhibitors , Factor XIa/metabolism , Anticoagulants/pharmacology , Blood Coagulation/drug effects , Male , Factor XIIa/antagonists & inhibitors , Factor XIIa/metabolism , Carotid Artery Thrombosis/prevention & control , Carotid Artery Thrombosis/chemically induced , Carotid Artery Thrombosis/drug therapy , Mice , Infarction, Middle Cerebral Artery/drug therapy , Hemorrhage/chemically induced , Mice, Inbred C57BL , Partial Thromboplastin Time
3.
Bioorg Chem ; 142: 106951, 2024 01.
Article in English | MEDLINE | ID: mdl-37924755

ABSTRACT

Thrombotic diseases, such as myocardial infarction, stroke, and deep vein thrombosis, severely threaten human health, and anticoagulation is an effective way to prevent such illnesses. However, most anticoagulant drugs in the clinic have different bleeding risks. Previous studies have shown that coagulation factor XI is an ideal target for safe anticoagulant drug development. Here, we designed the FXIa inhibitory peptide DX-88mut by replacing Loop1 (DGPCRAAHPR) and Loop2 (IYGGC) in DX-88, which is a clinical drug targeting PKa for the treatment of hereditary angioedema, using Loop1 (TGPCRAMISR) and Loop2 (FYGGC) in the FXIa inhibitory peptide PN2KPI, respectively. DX-88mut selectively inhibited FXIa against a panel of serine proteases with an IC50 value of 14.840 ± 0.453 nM, dose-dependently prolonged APTT in mouse, rat and human plasma, and potently inhibited FeCl3-induced carotid artery thrombosis in mice at a dose of 1 µmol/kg. Additionally, DX-88mut did not show a significant bleeding risk at a dose of 5 µmol/kg. Taken together, these results show that DX-88mut is a potential candidate for the development of a novel antithrombotic agent.


Subject(s)
Factor XIa , Thrombosis , Humans , Rats , Mice , Animals , Factor XIa/metabolism , Factor XIa/pharmacology , Blood Coagulation , Anticoagulants/pharmacology , Thrombosis/drug therapy , Peptides/pharmacology
4.
J Immunol ; 206(8): 1784-1792, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33811105

ABSTRACT

Complement factor H (CFH) is the major inhibitor of the alternative pathway of the complement system and is structurally related to beta2-glycoprotein I, which itself is known to bind to ligands, including coagulation factor XI (FXI). We observed reduced complement activation when FXI activation was inhibited in a baboon model of lethal systemic inflammation, suggesting cross-talk between FXI and the complement cascade. It is unknown whether FXI or its activated form, activated FXI (FXIa), directly interacts with the complement system. We explored whether FXI could interact with and inhibit the activity of CFH. We found that FXIa neutralized CFH by cleavage of the R341/R342 bonds. FXIa reduced the capacity of CFH to enhance the cleavage of C3b by factor I and the decay of C3bBb. The binding of CFH to human endothelial cells was also reduced after incubating CFH with FXIa. The addition of either short- or long-chain polyphosphate enhanced the capacity of FXIa to cleave CFH. FXIa also cleaved CFH that was present on endothelial cells and in the secretome from blood platelets. The generation of FXIa in plasma induced the cleavage of CFH. Moreover, FXIa reduced the cleavage of C3b by factor I in serum. Conversely, we observed that CFH inhibited FXI activation by either thrombin or FXIIa. Our study provides, to our knowledge, a novel molecular link between the contact pathway of coagulation and the complement system. These results suggest that FXIa generation enhances the activity of the complement system and thus may potentiate the immune response.


Subject(s)
Blood Platelets/metabolism , Complement Factor H/metabolism , Endothelial Cells/metabolism , Factor XIa/metabolism , Inflammation/metabolism , Animals , Blood Coagulation , Complement C3b/metabolism , Complement Pathway, Alternative , Fibrinogen/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Papio , Protein Binding , Receptor Cross-Talk
5.
Eur J Clin Invest ; 52(12): e13857, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35996895

ABSTRACT

BACKGROUND: Coronary artery disease (CAD) is associated with a prothrombotic tendency including increased factor (F) VIIa-antithrombin (FVIIa-AT) complexes, a measure of tissue factor (TF) exposure, and activated FXI (FXIa). We investigated whether increased FVIIa-AT complexes are associated with FXIa and active TF and if major adverse clinical outcomes are predicted by the complexes in CAD. METHODS: In 120 CAD patients, we assessed FVIIa-AT complex concentrations and the presence of circulating FXIa and active TF. Levels of 8-iso-prostaglandin F2α (8-iso-PGF2α), interleukin-6, high-sensitivity C reactive protein, prothrombin fragment 1 + 2, and free Tissue Factor Pathway Inhibitor were determined. Myocardial infarction (MI), ischemic stroke, systemic thromboembolism (SE), and cardiovascular (CV) death were recorded separately and as a composite endpoint, during follow-up. RESULTS: FVIIa-AT complexes were positively associated with current smoking and multivessel CAD. Elevated FVIIa-AT complexes characterized patients with circulating FXIa and/or active TF in association with increased plasma isoprostanes but not with thrombin generation or inflammatory markers. During a median follow-up of 106 months (interquartile range 95-119), high baseline levels of FVIIa-AT complexes predicted ischemic stroke/SE (HR 4.61 [95% CI 1.48-18.42]) and a composite endpoint of MI, stroke/SE, and CV death (HR 7.47 [95% CI 2.81-19.87]). CONCLUSIONS: This study is the first to show that high FVIIa-AT complexes characterize advanced CAD patients with detectable FXIa and active TF, which is, in part, driven by oxidative stress. High FVIIa-AT complexes were associated with the risk of ischemic stroke/SE during long-term follow-up, highlighting the need for effective antithrombotic agents in CAD.


Subject(s)
Coronary Artery Disease , Ischemic Stroke , Myocardial Infarction , Humans , Factor XIa/metabolism , Factor VIIa , Antithrombins , Antithrombin III , Thromboplastin/metabolism , Anticoagulants
6.
Am J Physiol Cell Physiol ; 320(3): C365-C374, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33471623

ABSTRACT

Factor XI (FXI) has been shown to bind platelets, but the functional significance of this observation remains unknown. Platelets are essential for hemostasis and play a critical role in thrombosis, whereas FXI is not essential for hemostasis but promotes thrombosis. An apparent functional contradiction, platelets are known to support thrombin generation, yet platelet granules release protease inhibitors, including those of activated FXI (FXIa). We aim to investigate the secretory and binding mechanisms by which platelets could support or inhibit FXIa activity. The presence of platelets enhanced FXIa activity in a purified system and increased coagulation Factor IX (FIX) activation by FXIa and fibrin generation in human plasma. In contrast, platelets reduced the activation of FXI by activated coagulation factor XII (FXIIa) and the activation of FXII by kallikrein (PKa). Incubation of FXIa with the platelet secretome, which contains FXIa inhibitors, such as protease nexin-II, abolished FXIa activity, yet in the presence of activated platelets, the secretome was not able to block the activity of FXIa. FXIa variants lacking the anion-binding sites did not alter the effect of platelets on FXIa activity or interaction. Western blot analysis of bound FXIa [by FXIa-platelet membrane immunoprecipitation] showed that the interaction with platelets is zinc dependent and, unlike FXI binding to platelets, not dependent on glycoprotein Ib. FXIa binding to the platelet membrane increases its capacity to activate FIX in plasma likely by protecting it from inhibition by inhibitors secreted by activated platelets. Our findings suggest that an interaction of FXIa with the platelet surface may induce an allosteric modulation of FXIa.


Subject(s)
Blood Platelets/metabolism , Factor XIa/metabolism , Adolescent , Amyloid beta-Protein Precursor/metabolism , Binding Sites/physiology , Blood Coagulation/physiology , Female , Hemostasis/physiology , Humans , Male , Thrombin/metabolism , Thrombosis/metabolism
7.
Blood ; 131(3): 353-364, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29158361

ABSTRACT

Chemerin is a chemoattractant and adipokine that circulates in blood as inactive prochemerin (chem163S). Chem163S is activated by a series of C-terminal proteolytic cleavages resulting in diverse chemerin forms with different levels of activity. We screened a panel of proteases in the coagulation, fibrinolytic, and inflammatory cascades to identify those that process prochemerin in plasma. Factor XIa (FXIa) cleaved chem163S, generating a novel chemerin form, chem162R, as an intermediate product, and chem158K, as the final product. Processing at Arg162 was not required for cleavage at Lys158 or regulation of chemerin bioactivity. Contact phase activation of human platelet-poor plasma by kaolin led to cleavage of chem163S, which was undetectable in FXI-depleted plasma and markedly enhanced in platelet-rich plasma (PRP). Contact phase activation by polyphosphate in PRP resulted in 75% cleavage of chem163S. This cleavage was partially inhibited by hirudin, which blocks thrombin activation of FXI. After activation of plasma, levels of the most potent form of chemerin, chem157S, as well as inactive chem155A, increased. Plasma levels of chem163S in FXI-deficient patients were significantly higher compared with a matched control group (91 ± 10 ng/mL vs 58 ± 3 ng/mL, n = 8; P < .01) and inversely correlated with the plasma FXI levels. Thus FXIa, generated on contact phase activation, cleaves chem163S to generate chem158K, which can be further processed to the most active chemerin form, providing a molecular link between coagulation and inflammation.


Subject(s)
Blood Coagulation , Chemokines/metabolism , Factor XIa/metabolism , Inflammation/pathology , Intercellular Signaling Peptides and Proteins/metabolism , Amino Acid Sequence , Arginine/metabolism , Blood Platelets/metabolism , Cell-Derived Microparticles/metabolism , Chemokines/blood , Chemokines/chemistry , Humans , Hydrolysis , Intercellular Signaling Peptides and Proteins/blood , Intercellular Signaling Peptides and Proteins/chemistry , Kinetics , Peptides/chemistry , Peptides/metabolism , Phospholipids/metabolism , Protein Isoforms/blood
8.
PLoS Comput Biol ; 15(8): e1007266, 2019 08.
Article in English | MEDLINE | ID: mdl-31381558

ABSTRACT

During thrombosis, thrombin generates fibrin, however fibrin reversibly binds thrombin with low affinity E-domain sites (KD = 2.8 µM) and high affinity γ'-fibrin sites (KD = 0.1 µM). For blood clotting on collagen/tissue factor (1 TF-molecule/µm2) at 200 s-1 wall shear rate, high µM-levels of intraclot thrombin suggest robust prothrombin penetration into clots. Setting intraclot zymogen concentrations to plasma levels (and neglecting cofactor rate limitations) allowed the linearization of 7 Michaelis-Menton reactions between 6 species to simulate intraclot generation of: Factors FXa (via TF/VIIa or FIXa), FIXa (via TF/FVIIa or FXIa), thrombin, fibrin, and FXIa. This reduced model [7 rates, 2 KD's, enzyme half-lives~1 min] predicted the measured clot elution rate of thrombin-antithrombin (TAT) and fragment F1.2 in the presence and absence of the fibrin inhibitor Gly-Pro-Arg-Pro. To predict intraclot fibrin reaching 30 mg/mL by 15 min, the model required fibrinogen penetration into the clot to be strongly diffusion-limited (actual rate/ideal rate = 0.05). The model required free thrombin in the clot (~100 nM) to have an elution half-life of ~2 sec, consistent with measured albumin elution, with most thrombin (>99%) being fibrin-bound. Thrombin-feedback activation of FXIa became prominent and reached 5 pM FXIa at >500 sec in the simulation, consistent with anti-FXIa experiments. In predicting intrathrombus thrombin and fibrin during 15-min microfluidic experiments, the model revealed "cascade amplification" from 30 pM levels of intrinsic tenase to 15 nM prothrombinase to 15 µM thrombin to 90 µM fibrin. Especially useful for multiscale simulation, this reduced model predicts thrombin and fibrin co-regulation during thrombosis under flow.


Subject(s)
Blood Coagulation/physiology , Models, Biological , Thrombosis/blood , Blood Platelets/metabolism , Collagen/blood , Computational Biology , Computer Simulation , Cysteine Endopeptidases/blood , Factor XIa/metabolism , Fibrin/metabolism , Humans , In Vitro Techniques , Kinetics , Neoplasm Proteins/blood , Regional Blood Flow/physiology , Thrombin/metabolism , Thromboplastin/metabolism
9.
Bioorg Med Chem Lett ; 30(16): 127072, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32340773

ABSTRACT

A series of 4, 4-disubstituted proline analogs were designed, synthesized, and tested for selective inhibition of blood coagulation factor XIa in search of new non-vitamin K antagonists based oral anticoagulants for potential prevention and treatment of thrombotic diseases. Starting from a potent thrombin (FIIa) inhibitor chemotype with FIIa IC50 = 1 nM and FXIa IC50 = 160 nM, medicinal chemistry iterations guided by molecular modeling and structure-based drug design led to steady improvement of FXIa potency while dialing down thrombin activity and improving selectivity. Through this exercise, a thousand-fold enhancement of selectivity over thrombin was achieved with some analogs carrying factor XIa inhibition potencies in the 10 nM range. In this communication, we discuss the design principles and structure activity relationship (SAR) of these novel FXIa selective inhibitors.


Subject(s)
Anticoagulants/pharmacology , Drug Design , Factor XIa/antagonists & inhibitors , Proline/pharmacology , Anticoagulants/chemical synthesis , Anticoagulants/chemistry , Dose-Response Relationship, Drug , Factor XIa/metabolism , Humans , Molecular Structure , Proline/chemical synthesis , Proline/chemistry , Structure-Activity Relationship
10.
Bioorg Med Chem Lett ; 30(4): 126949, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31932224

ABSTRACT

The discovery of orally bioavailable FXIa inhibitors has been a challenge. Herein, we describe our efforts to address this challenge by optimization of our imidazole-based macrocyclic series. Our optimization strategy focused on modifications to the P2 prime, macrocyclic amide linker, and the imidazole scaffold. Replacing the amide of the macrocyclic linker with amide isosteres led to the discovery of substituted amine linkers which not only maintained FXIa binding affinity but also improved oral exposure in rats. Combining the optimized macrocyclic amine linker with a pyridine scaffold afforded compounds 23 and 24 that were orally bioavailable, single-digit nanomolar FXIa inhibitors with excellent selectivity against relevant blood coagulation enzymes.


Subject(s)
Amines/chemistry , Factor XIa/antagonists & inhibitors , Macrocyclic Compounds/chemistry , Serine Proteinase Inhibitors/chemical synthesis , Administration, Oral , Animals , Binding Sites , Drug Design , Factor XIa/metabolism , Half-Life , Macrocyclic Compounds/metabolism , Macrocyclic Compounds/pharmacokinetics , Molecular Dynamics Simulation , Protein Structure, Tertiary , Pyridines/chemistry , Rats , Serine Proteinase Inhibitors/metabolism , Serine Proteinase Inhibitors/pharmacokinetics , Structure-Activity Relationship
11.
Bioorg Med Chem ; 28(23): 115762, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32992249

ABSTRACT

Human factor XIa (hFXIa) has emerged as an attractive target for development of new anticoagulants that promise higher level of safety. Different strategies have been adopted so far for the design of anti-hFXIa molecules including competitive and non-competitive inhibition. Of these, allosteric dysfunction of hFXIa's active site is especially promising because of the possibility of controlled reduction in activity that may offer a route to safer anticoagulants. In this work, we assess fragment-based design approach to realize a group of novel allosteric hFXIa inhibitors. Starting with our earlier discovery that sulfated quinazolinone (QAO) bind in the heparin-binding site of hFXIa, we developed a group of two dozen dimeric sulfated QAOs with intervening linkers that displayed a progressive variation in inhibition potency. In direct opposition to the traditional wisdom, increasing linker flexibility led to higher potency, which could be explained by computational studies. Sulfated QAO 19S was identified as the most potent and selective inhibitor of hFXIa. Enzyme inhibition studies revealed that 19S utilizes a non-competitive mechanism of action, which was supported by fluorescence studies showing a classic sigmoidal binding profile. Studies with selected mutants of hFXIa indicated that sulfated QAOs bind in heparin-binding site of the catalytic domain of hFXIa. Overall, the approach of fragment-based design offers considerable promise for designing heparin-binding site-directed allosteric inhibitors of hFXIa.


Subject(s)
Drug Design , Factor XIa/antagonists & inhibitors , Serine Proteinase Inhibitors/chemistry , Allosteric Regulation/drug effects , Binding Sites , Catalytic Domain , Dimerization , Factor XIa/metabolism , Humans , Kinetics , Molecular Docking Simulation , Quinazolinones/chemistry , Quinazolinones/metabolism , Quinazolinones/pharmacology , Serine Proteinase Inhibitors/metabolism , Structure-Activity Relationship , Sulfates/chemistry
12.
Immunol Rev ; 274(1): 281-289, 2016 11.
Article in English | MEDLINE | ID: mdl-27782339

ABSTRACT

The blood plasma contains four biologically important proteolytic cascades, which probably evolved from the same ancestral gene. This in part may explain why each cascade has very similar "initiating trigger" followed by sequential and cascade-like downstream enzymatic activation pattern. The four cascades are: the complement system, the blood clotting cascade, the fibrinolytic system, and the kallikrein-kinin system. Although much has been written about the interplay between all these enzymatic cascades, the cross-talk between the complement and the kinin generating systems has become particularly relevant as this interaction results in the generation of nascent molecules that have significant impact in various inflammatory diseases including angioedema and cancer. In this review, we will focus on the consequences of the interplay between the two systems by highlighting the role of a novel molecular link called gC1qR. Although this protein was first identified as a receptor for C1q, it is now recognized as a multiligand binding cellular protein, which serves not only as C1q receptor, but also as high affinity (KD  ≤ 0.8 nM) binding site for both high molecular weight kininogen (HK) and factor XII (FXII). At inflammatory sites, where atherogenic factors such as immune complexes and/or pathogens can activate the endothelial cell into a procoagulant and proinflammatory surface, the two pathways are activated to generate vasoactive peptides that contribute in various ways to the inflammatory processes associated with numerous diseases. More importantly, since recent observations strongly suggest an important role for both pathways in cancer, we will focus on how a growing tumor cluster can employ the byproducts derived from the two activation systems to ensure not only its survival and growth, but also its escape into distal sites of colonization.


Subject(s)
Angioedema/immunology , Atherosclerosis/immunology , Complement C1q/metabolism , Factor XIa/metabolism , Immunity, Innate , Inflammation/immunology , Neoplasms/immunology , Animals , Humans , Proteolysis , Receptor Cross-Talk , Signal Transduction
13.
Am J Physiol Cell Physiol ; 316(3): C377-C392, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30566391

ABSTRACT

Activated coagulation factor XI (FXIa) is a serine proteinase that plays a key role in the intrinsic coagulation pathway. The analysis of FXI-knockout mice has indicated the contribution of FXI to the pathogenesis of atherosclerosis. However, the underlying mechanism remains unknown. We hypothesized that FXIa exerts vascular smooth muscle effects via proteinase-activated receptor 1 (PAR1). Fura-2 fluorometry revealed that FXIa elicited intracellular Ca2+ signal in rat embryo aorta smooth muscle A7r5 cells. The influx of extracellular Ca2+ played a greater role in generating Ca2+ signal than the Ca2+ release from intracellular stores. The FXIa-induced Ca2+ signal was abolished by the pretreatment with atopaxar, an antagonist of PAR1, or 4-amidinophenylmethanesulfonyl fluoride (p-APMSF), an inhibitor of proteinase, while it was also lost in embryonic fibroblasts derived from PAR1-/- mice. FXIa cleaved the recombinant protein containing the extracellular region of PAR1 at the same site (R45/S46) as that of thrombin, a canonical PAR1 agonist. The FXIa-induced Ca2+ influx was inhibited by diltiazem, an L-type Ca2+ channel blocker, and by siRNA targeted to CaV1.2. The FXIa-induced Ca2+ influx was also inhibited by GF109203X and rottlerin, inhibitors of protein kinase C. In a wound healing assay, FXIa increased the rate of cell migration by 2.46-fold of control, which was partly inhibited by atopaxar or diltiazem. In conclusion, FXIa mainly elicits the Ca2+ signal via the PAR1/CaV1.2-mediated Ca2+ influx and accelerates the migration in vascular smooth muscle cells. The present study provides the first evidence that FXIa exerts a direct cellular effect on vascular smooth muscle.


Subject(s)
Calcium/metabolism , Cell Movement/physiology , Factor XIa/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Blood Coagulation/physiology , Calcium Channels, L-Type/metabolism , Cell Line , Female , Kinetics , Male , Mice , Mice, Inbred C57BL , Pregnancy , Protein Binding/physiology , Rats , Rats, Wistar , Thrombin/metabolism
14.
Blood ; 130(4): 527-536, 2017 07 27.
Article in English | MEDLINE | ID: mdl-28611024

ABSTRACT

The amyloid precursor protein (APP), primarily known as the precursor of amyloid peptides that accumulate in the brain of patients with Alzheimer disease, is abundant in platelets, but its physiological function remains unknown. In this study, we investigated the role of APP in hemostasis and thrombosis, using APP knockout (KO) mice. Ex vivo aggregation, secretion, and integrin αIIbß3 inside-out activation induced by several agonists were normal in APP-deficient platelets, but the number of circulating platelets was reduced by about 20%, and their size was slightly increased. Tail bleeding time was normal, and in vivo, the absence of APP did not alter thrombus formation in the femoral artery. In contrast, in a model of vein thrombosis induced by flow restriction in the inferior vena cava, APP-KO mice, as well as chimeric mice with selective deficiency of APP in blood cells, developed much larger thrombi than control animals, and were more sensitive to embolization. Consistent with this, in a pulmonary thromboembolism model, larger vessels were occluded. APP-KO mice displayed a shorter APTT, but not PT, when measured in the presence of platelets. Moreover, the activity of factor XIa (FXIa), but not FXIIa, was higher in APP-KO mice compared with controls. APP-KO mice presented a higher number of circulating platelet-leukocyte aggregates, and neutrophils displayed a greater tendency to protrude extracellular traps, which were more strongly incorporated into venous thrombi. These results indicate that platelet APP limits venous thromboembolism through a negative regulation of both fibrin formation and neutrophil function.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Blood Platelets/metabolism , Vena Cava, Inferior/metabolism , Venous Thromboembolism/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Blood Platelets/pathology , Factor XIa/genetics , Factor XIa/metabolism , Mice , Mice, Knockout , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Vena Cava, Inferior/pathology , Venous Thromboembolism/genetics , Venous Thromboembolism/pathology
15.
Haemophilia ; 24(5): 815-822, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30112856

ABSTRACT

INTRODUCTION: The thrombin generation assay (TGA) can be used to monitor factor replacement therapy in patients with haemophilia. The TGA assay is typically performed using tissue factor as the reaction activator; however, activating with FIXa or FXIa can enhance assay sensitivity when FVIII < 1%. AIMS: To evaluate the sensitivity of the TGA when FIXa (5 nmol/L) and FXIa (0.22 nmol/L) are used to activate the assay in platelet-poor plasma and to compare these data to the one-stage and chromogenic assays. METHODS: Plasma from 10 severe FVIII-deficient subjects was supplemented with FVIII (0%, 0.1%, 0.4%, 1.2%, 4%, 11% and 33%), using either Novo Eight® , Advate® , Eloctate® , turoctocog alfa pegol or a control standard. The one-stage and chromogenic assays quantified the FVIII levels. The TGA assay was activated using either FIXa or FXIa. RESULTS: Both FIXa- and FXIa-activated TGA were sensitive across FVIII concentrations, with intra-assay coefficient of variation (CV) < 10%. The FXIa-activated assay had 25% CV at the lowest level of FVIII compared to 10% CV with FIXa activation. There were strong correlations between the FIXa- and FXIa-activated TGA tests (R2  = 0.9912) and between the one-stage and chromogenic assays (R2  = 0.9469). However, there were poor relationships between the TGA tests and one-stage and chromogenic assays. CONCLUSIONS: Both FIXa- and FXIa activation results in similar TGA profiles across a FVIII range of 0.1%-33%; however, FIXa activation was more robust at the lowest levels of FVIII compared with FXIa activation.


Subject(s)
Factor VIII/metabolism , Factor XIa/metabolism , Hemophilia A/blood , Thrombin/metabolism , Humans
16.
J Org Chem ; 83(10): 5398-5409, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29667397

ABSTRACT

Silanediol peptidomimetics have been prepared, designed to inhibit the serine protease enzyme Factor XIa (FXIa) for treatment of thrombosis without complete interruption of normal hemostasis. These Arg-[Si]-Ala analogues of the FXIa substrate (FIX) are the first silanediol dipeptide analogues to carry a basic guanidine group. Control of stereochemistry was accomplished using catalytic asymmetric hydrosilylation and addition of a silyllithium intermediate to the Davis-Ellman sulfinimine.


Subject(s)
Factor XIa/antagonists & inhibitors , Serine Proteinase Inhibitors/pharmacology , Silanes/pharmacology , Factor XIa/metabolism , Humans , Molecular Conformation , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/chemistry , Silanes/chemical synthesis , Silanes/chemistry
18.
Blood ; 126(12): 1403-4, 2015 Sep 17.
Article in English | MEDLINE | ID: mdl-26384283

ABSTRACT

In this issue of Blood, Zhu et al have established, in human blood, that factor XIa and polyphosphate make significant contributions to thrombus formation. This makes these molecules good targets for therapeutic intervention.


Subject(s)
Blood Coagulation , Blood Platelets/metabolism , Factor XIa/metabolism , Polyphosphates/metabolism , Thromboplastin/metabolism , Humans
19.
Blood ; 125(9): 1488-96, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25587039

ABSTRACT

Activation of coagulation factor XI (FXI) may play a role in hemostasis. The primary substrate of activated FXI (FXIa) is FIX, leading to FX activation (FXa) and thrombin generation. However, recent studies suggest the hemostatic role of FXI may not be restricted to the activation of FIX. We explored whether FXI could interact with and inhibit the activity of tissue factor pathway inhibitor (TFPI). TFPI is an essential reversible inhibitor of activated factor X (FXa) and also inhibits the FVIIa-TF complex. We found that FXIa neutralized both endothelium- and platelet-derived TFPI by cleaving the protein between the Kunitz (K) 1 and K2 domains (Lys86/Thr87) and at the active sites of the K2 (Arg107/Gly108) and K3 (Arg199/Ala200) domains. Addition of FXIa to plasma was able to reverse the ability of TFPI to prolong TF-initiated clotting times in FXI- or FIX-deficient plasma, as well as FXa-initiated clotting times in FX-deficient plasma. Treatment of cultured endothelial cells with FXIa increased the generation of FXa and promoted TF-dependent fibrin formation in recalcified plasma. Together, these results suggest that the hemostatic role of FXIa may be attributed not only to activation of FIX but also to promoting the extrinsic pathway of thrombin generation through inactivation of TFPI.


Subject(s)
Blood Coagulation/physiology , Blood Platelets/metabolism , Factor IX/metabolism , Factor XIa/metabolism , Factor Xa/metabolism , Fibrin/metabolism , Lipoproteins/metabolism , Blood Platelets/cytology , Blotting, Western , Cells, Cultured , Flow Cytometry , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Lipoproteins/genetics , Mutation/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
20.
Blood ; 126(12): 1494-502, 2015 Sep 17.
Article in English | MEDLINE | ID: mdl-26136249

ABSTRACT

Factor XIIa (FXIIa) and factor XIa (FXIa) contribute to thrombosis in animal models, whereas platelet-derived polyphosphate (polyP) may potentiate contact or thrombin-feedback pathways. The significance of these mediators in human blood under thrombotic flow conditions on tissue factor (TF) -bearing surfaces remains inadequately resolved. Human blood (corn trypsin inhibitor treated [4 µg/mL]) was tested by microfluidic assay for clotting on collagen/TF at TF surface concentration ([TF]wall) from ∼0.1 to 2 molecules per µm(2). Anti-FXI antibodies (14E11 and O1A6) or polyP-binding protein (PPXbd) were used to block FXIIa-dependent FXI activation, FXIa-dependent factor IX (FIX) activation, or platelet-derived polyP, respectively. Fibrin formation was sensitive to 14E11 at 0 to 0.1 molecules per µm(2) and sensitive to O1A6 at 0 to 0.2 molecules per µm(2). However, neither antibody reduced fibrin generation at ∼2 molecules per µm(2) when the extrinsic pathway became dominant. Interestingly, PPXbd reduced fibrin generation at low [TF]wall (0.1 molecules per µm(2)) but not at zero or high [TF]wall, suggesting a role for polyP distinct from FXIIa activation and requiring low extrinsic pathway participation. Regardless of [TF]wall, PPXbd enhanced fibrin sensitivity to tissue plasminogen activator and promoted clot retraction during fibrinolysis concomitant with an observed PPXbd-mediated reduction of fibrin fiber diameter. This is the first detection of endogenous polyP function in human blood under thrombotic flow conditions. When triggered by low [TF]wall, thrombosis may be druggable by contact pathway inhibition, although thrombolytic susceptibility may benefit from polyP antagonism regardless of [TF]wall.


Subject(s)
Blood Coagulation , Blood Platelets/metabolism , Factor XIa/metabolism , Polyphosphates/metabolism , Thromboplastin/metabolism , Antibodies/pharmacology , Blood Coagulation Tests , Blood Platelets/cytology , Collagen/metabolism , Humans , Molecular Targeted Therapy , Signal Transduction , Thrombin/metabolism , Thrombosis/drug therapy , Thrombosis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL