Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Med Chem ; 63(15): 8088-8113, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32551603

ABSTRACT

The serine protease factor XI (FXI) is a prominent drug target as it holds promise to deliver efficacious anticoagulation without an enhanced risk of major bleeds. Several efforts have been described targeting the active form of the enzyme, FXIa. Herein, we disclose our efforts to identify potent, selective, and orally bioavailable inhibitors of FXIa. Compound 1, identified from a diverse library of internal serine protease inhibitors, was originally designed as a complement factor D inhibitor and exhibited submicromolar FXIa activity and an encouraging absorption, distribution, metabolism, and excretion (ADME) profile while being devoid of a peptidomimetic architecture. Optimization of interactions in the S1, S1ß, and S1' pockets of FXIa through a combination of structure-based drug design and traditional medicinal chemistry led to the discovery of compound 23 with subnanomolar potency on FXIa, enhanced selectivity over other coagulation proteases, and a preclinical pharmacokinetics (PK) profile consistent with bid dosing in patients.


Subject(s)
Factor XIa/antagonists & inhibitors , Factor XIa/genetics , Factor Xa Inhibitors/administration & dosage , Factor Xa Inhibitors/chemistry , Administration, Oral , Amino Acid Sequence , Animals , Biological Availability , Dogs , Drug Evaluation, Preclinical/methods , Humans , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
2.
J Thromb Haemost ; 17(12): 2047-2055, 2019 12.
Article in English | MEDLINE | ID: mdl-31519061

ABSTRACT

BACKGROUND: Factor XI (FXI) is a zymogen in the coagulation pathway that, once activated, promotes haemostasis by activating factor IX (FIX). Substitution studies using apple domains of the homologous protein prekallikrein have identified that FIX binds to the apple 3 domain of FXI. However, the molecular changes upon activation of FXI or binding of FIX to FXIa have remained largely unresolved. OBJECTIVES: This study aimed to gain more insight in the FXI activation mechanism by identifying the molecular differences between FXI and FXIa, and in the conformational changes in FXIa induced by binding of FIX. METHODS: Hydrogen-deuterium exchange mass spectrometry was performed on FXI, FXIa, and FXIa in complex with FIX. RESULTS: Both activation and binding to FIX induced conformational changes at the interface between the catalytic domain and the apple domains of FXI(a)-more specifically at the loops connecting the apple domains. Moreover, introduction of FIX uniquely induced a reduction of deuterium uptake in the beginning of the apple 3 domain. CONCLUSIONS: We propose that the conformational changes of the catalytic domain upon activation increase the accessibility to the apple 3 domain to enable FIX binding. Moreover, our HDX MS results support the location of the proposed FIX binding site at the beginning of the apple 3 domain and suggest a mediating role in FIX binding for both loops adjacent to the apple 3 domain.


Subject(s)
Factor IX/metabolism , Factor XI/metabolism , Factor XIa/metabolism , Hemostasis , Hydrogen Deuterium Exchange-Mass Spectrometry , Enzyme Activation , Factor IX/chemistry , Factor XI/chemistry , Factor XI/genetics , Factor XIa/chemistry , Factor XIa/genetics , HEK293 Cells , Humans , Models, Molecular , Protein Binding , Protein Interaction Domains and Motifs , Structure-Activity Relationship
3.
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
4.
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
5.
J Thromb Haemost ; 14(12): 2509-2523, 2016 12.
Article in English | MEDLINE | ID: mdl-27797450

ABSTRACT

Essentials Current antifibrinolytics - aminocaproic acid and tranexamic acid-can cause seizures or renal injury. KD1L17R -KT , aprotinin and tranexamic acid were tested in a modified mouse tail-amputation model. S2'-subsite variations between human and mouse factor XIa result in vastly different inhibition profiles. KD1L17R -KT reduces blood loss and D-dimer levels in mouse with unobserved seizures or renal injury. SUMMARY: Background Using tissue factor pathway inhibitor (TFPI)-2 Kunitz domain1 (KD1), we obtained a bifunctional antifibrinolytic molecule (KD1L17R -KT ) with C-terminal lysine (kringle domain binding) and P2'-residue arginine (improved specificity towards plasmin). KD1L17R -KT strongly inhibited human plasmin (hPm), with no inhibition of human kallikrein (hKLK) or factor XIa (hXIa). Furthermore, KD1L17R -KT reduced blood loss comparable to aprotinin in a mouse liver-laceration model of organ hemorrhage. However, effectiveness of these antifibrinolytic agents in a model of hemorrhage mimicking extremity trauma and their inhibition efficiencies for mouse enzymes (mPm, mKLK or mXIa) remain to be determined. Objective To determine potential differences in inhibition constants of various antifibrinolytic agents against mouse and human enzymes and test their effectiveness in a modified mouse tail-amputation hemorrhage model. Methods/Results Unexpectedly, mXIa was inhibited with ~ 17-fold increased affinity by aprotinin (Ki ~ 20 nm) and with measurable affinity for KD1L17R -KT (Ki ~ 3 µm); in contrast, KD1WT -VT inhibited hXIa or mXIa with similar affinity. Compared with hPm, mPm had ~ 3-fold reduced affinity, whereas species specificity for hKLK and mKLK was comparable for each inhibitor. S2'-subsite variations largely accounted for the observed differences. KD1L17R -KT and aprotinin were more effective than KD1WT -VT or tranexamic acid in inhibiting tPA-induced mouse plasma clot lysis. Further, KD1L17R -KT was more effective than KD1WT -VT and was comparable to aprotinin and tranexamic acid in reducing blood loss and D-dimer levels in the mouse tail-amputation model. Conclusions Inhibitor potencies differ between antifibrinolytic agents against human and mouse enzymes. KD1L17R -KT is effective in reducing blood loss in a tail-amputation model that mimics extremity injury.


Subject(s)
Factor XIa/genetics , Fibrinolysin/genetics , Glycoproteins/chemistry , Kallikreins/genetics , Animals , Antifibrinolytic Agents , Aprotinin/chemistry , Cattle , Fibrin Fibrinogen Degradation Products/chemistry , Fibrinolysis , Glycoproteins/genetics , Hemorrhage , Humans , Leucine/chemistry , Liver/metabolism , Mice , Models, Molecular , Mutation , Peptide Hydrolases/chemistry , Protein Domains , Seizures , Species Specificity , Tranexamic Acid/chemistry , Trypsin/chemistry
6.
PLoS One ; 8(8): e74043, 2013.
Article in English | MEDLINE | ID: mdl-24013493

ABSTRACT

Hereditary angioedema (HAE) is predominantly caused by a deficiency in C1 esterase inhibitor (C1INH) (HAE-C1INH). C1INH inhibits activated factor XII (FXIIa), activated factor XI (FXIa), and kallikrein. In HAE-C1INH patients the thrombotic risk is not increased even though activation of the contact system is poorly regulated. Therefore, we hypothesized that contact activation preferentially leads to kallikrein formation and less to activation of the coagulation cascade in HAE-C1INH patients. We measured the levels of C1INH in complex with activated contact factors in plasma samples of HAE-C1INH patients (N=30, 17 during remission and 13 during acute attack) and healthy controls (N=10). We did not detect differences in enzyme-inhibitor complexes between samples of controls, patients during remission and patients during an acute attack. Reconstitution with C1INH did not change this result. Next, we determined the potential to form enzyme-inhibitory complexes after complete in vitro activation of the plasma samples with a FXII trigger. In all samples, enzyme-C1INH levels increased after activation even in patients during an acute attack. However, the levels of FXIIa-C1INH, FXIa-C1INH and kallikrein-C1INH were at least 52% lower in samples taken during remission and 70% lower in samples taken during attack compared to samples from controls (p<0.05). Addition of C1INH after activation led to an increase in levels of FXIIa-C1INH and FXIa-C1INH (p<0.05), which were still lower than in controls (p<0.05), while the levels of kallikrein-C1INH did not change. These results are consistent with constitutive activation and attenuated depletion of the contact system and show that the ongoing activation of the contact system, which is present in HAE-C1INH patients both during remission and during acute attacks, is not associated with preferential generation of kallikrein over FXIa.


Subject(s)
Angioedemas, Hereditary/blood , Complement C1 Inhibitor Protein/metabolism , Factor XIIa/metabolism , Factor XIa/metabolism , Kallikreins/blood , Multiprotein Complexes/blood , Adult , Aged , Angioedemas, Hereditary/genetics , Complement C1 Inhibitor Protein/genetics , Factor XIIa/genetics , Factor XIa/genetics , Female , Humans , Kallikreins/genetics , Male , Middle Aged , Multiprotein Complexes/genetics
7.
J Thromb Haemost ; 11(7): 1374-84, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23617568

ABSTRACT

BACKGROUND: A patient with factor XI (FXI) deficiency was reported with an Arg184Gly substitution in the FXI A3 domain. The A3 domain contains an exosite required for binding of FIX to activated FXI (FXIa). OBJECTIVE: To test the effects of the Arg184Gly substitution on FIX activation, and to characterize the FIX-binding site on FXIa. METHODS: Recombinant FXIa and FIX variants were used to identify residues involved in FIX activation by FXIa. Analysis of the FXI structure was used to identify potential FIX-binding sites. RESULTS: The Km for FIX activation by FXIa-Gly184 was approximately three-fold higher than for FXIa, suggesting that Arg184 is part of the exosite. Arg184 and the adjacent residues, Ile183 and Asp185, contribute to charged and hydrophobic areas that are not present in the FXI homolog prekallikrein (PK). Replacing residues 183-185 with alanine abolished exosite activity, similarly to replacement of the entire A3 domain with the A3 domain from PK (FXIa/PKA3). Reintroducing FXI residues 183-185 into FXIa/PKA3 partially restored the exosite, and replacing residues 183-185 and 260-264 completely restored exosite function. FIX in which the Ω-loop (residues 4-11) was replaced with the FVII Ω-loop was activated poorly by FXIa, suggesting that the FIX Ω-loop binds to FXIa. CONCLUSIONS: The results support a model in which the Ω-loop of FIX binds to an area on FXIa composed of residues from the N-terminus and C-terminus of the A3 domain. These residues are buried in zymogen FXI, and must be exposed upon conversion to FXIa to permit FIX binding.


Subject(s)
Blood Coagulation , Factor IX/metabolism , Factor XI Deficiency/blood , Factor XIa/metabolism , Arginine , Blood Coagulation Tests , Enzyme Activation , Factor IX/chemistry , Factor IX/genetics , Factor XI Deficiency/genetics , Factor XIa/chemistry , Factor XIa/genetics , Glycine , HEK293 Cells , Humans , Kinetics , Models, Molecular , Mutation , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Structure-Activity Relationship , Transfection
8.
J Biol Chem ; 287(45): 38200-9, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-22961984

ABSTRACT

During blood coagulation, the protease factor XIa (fXIa) activates factor IX (fIX). We describe a new mechanism for this process. FIX is cleaved initially after Arg(145) to form fIXα, and then after Arg(180) to form the protease fIXaß. FIXα is released from fXIa, and must rebind for cleavage after Arg(180) to occur. Catalytic efficiency of cleavage after Arg(180) is 7-fold greater than for cleavage after Arg(145), limiting fIXα accumulation. FXIa contains four apple domains (A1-A4) and a catalytic domain. Exosite(s) on fXIa are required for fIX binding, however, there is lack of consensus on their location(s), with sites on the A2, A3, and catalytic domains described. Replacing the A3 domain with the prekallikrein A3 domain increases K(m) for fIX cleavage after Arg(145) and Arg(180) 25- and ≥ 90-fold, respectively, and markedly decreases k(cat) for cleavage after Arg(180). Similar results were obtained with the isolated fXIa catalytic domain, or fXIa in the absence of Ca(2+). Forms of fXIa lacking the A3 domain exhibit 15-fold lower catalytic efficiency for cleavage after Arg(180) than for cleavage after Arg(145), resulting in fIXα accumulation. Replacing the A2 domain does not affect fIX activation. The results demonstrate that fXIa activates fIX by an exosite- and Ca(2+)-mediated release-rebind mechanism in which efficiency of the second cleavage is enhanced by conformational changes resulting from the first cleavage. Initial binding of fIX and fIXα requires an exosite on the fXIa A3 domain, but not the A2 or catalytic domain.


Subject(s)
Factor IX/metabolism , Factor IXa/metabolism , Factor XIa/metabolism , Arginine/metabolism , Binding Sites/genetics , Binding, Competitive , Biocatalysis/drug effects , Calcium/metabolism , Calcium/pharmacology , Catalytic Domain , Electrophoresis, Polyacrylamide Gel , Factor XIa/chemistry , Factor XIa/genetics , HEK293 Cells , Humans , Kinetics , Mutation , Oligopeptides/metabolism , Protein Multimerization , Proteolysis , Pyrrolidonecarboxylic Acid/analogs & derivatives , Pyrrolidonecarboxylic Acid/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Substrate Specificity
9.
J Biol Chem ; 287(9): 6187-95, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22207756

ABSTRACT

In the intrinsic pathway of blood coagulation factor XIa (FXIa) activates factor IX (FIX) by cleaving the zymogen at Arg(145)-Ala(146) and Arg(180)-Val(181) bonds releasing an 11-kDa activation peptide. FXIa and its isolated light chain (FXIa-LC) cleave S-2366 at comparable rates, but FXIa-LC is a very poor activator of FIX, possibly because FIX undergoes allosteric modification on binding to an exosite on the heavy chain of FXIa (FXIa-HC) required for optimal cleavage rates of the two scissile bonds of FIX. However preincubation of FIX with a saturating concentration of isolated FXIa-HC did not result in any potentiation in the rate of FIX cleavage by FXIa-LC. Furthermore, if FIX binding via the heavy chain exosite of FXIa determines the affinity of the enzyme-substrate interaction, then the isolated FXIa-HC should inhibit the rate of FIX activation by depleting the substrate. However, whereas FXIa/S557A inhibited FIX activation of by FXIa, FXIa-HC did not. Therefore, we examined FIX binding to FXIa/S557A, FXIa-HC, FXIa-LC, FXIa/C362S/C482S, and FXIa/S557A/C362S/C482S. The heavy and light chains are disulfide-linked in FXIa/S557A but not in FXIa/C362S/C482S and FXIa/S557A/C362S/C482S. In an ELISA assay only FXI/S557A ligated FIX with high affinity. Partial reduction of FXIa/S557A to produce heavy and light chains resulted in decreased FIX binding, and this function was regained upon reformation of the disulfide linkage between the heavy and the light chains. We therefore conclude that substrate recognition by the FXIa exosite(s) requires disulfide-linked heavy and light chains.


Subject(s)
Blood Coagulation/physiology , Disulfides/chemistry , Factor IX/chemistry , Factor IX/metabolism , Factor XIa/chemistry , Factor XIa/metabolism , Binding, Competitive/physiology , Catalytic Domain/physiology , Enzyme Activation/physiology , Factor IX/genetics , Factor XIa/genetics , HEK293 Cells , Humans , Mutagenesis, Site-Directed , Oxidation-Reduction , Peptide Hydrolases/metabolism , Substrate Specificity/physiology
10.
J Biol Chem ; 286(11): 8740-51, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21220417

ABSTRACT

Protein Z-dependent protease inhibitor (ZPI) is a recently identified member of the serpin superfamily that functions as a cofactor-dependent regulator of blood coagulation factors Xa and XIa. Here we provide evidence that, in addition to the established cofactors, protein Z, lipid, and calcium, heparin is an important cofactor of ZPI anticoagulant function. Heparin produced 20-100-fold accelerations of ZPI reactions with factor Xa and factor XIa to yield second order rate constants approaching the physiologically significant diffusion limit (k(a) = 10(6) to 10(7) M(-1) s(-1)). The dependence of heparin accelerating effects on heparin concentration was bell-shaped for ZPI reactions with both factors Xa and XIa, consistent with a template-bridging mechanism of heparin rate enhancement. Maximal accelerations of ZPI-factor Xa reactions required calcium, which augmented the heparin acceleration by relieving Gla domain inhibition as previously shown for heparin bridging of the antithrombin-factor Xa reaction. Heparin acceleration of both ZPI-protease reactions was optimal at heparin concentrations and heparin chain lengths comparable with those that produce physiologically significant rate enhancements of other serpin-protease reactions. Protein Z binding to ZPI minimally affected heparin rate enhancements, indicating that heparin binds to a distinct site on ZPI and activates ZPI in its physiologically relevant complex with protein Z. Taken together, these results suggest that whereas protein Z, lipid, and calcium cofactors promote ZPI inhibition of membrane-associated factor Xa, heparin activates ZPI to inhibit free factor Xa as well as factor XIa and therefore may play a physiologically and pharmacologically important role in ZPI anticoagulant function.


Subject(s)
Blood Proteins/antagonists & inhibitors , Fibrinolytic Agents/metabolism , Heparin/metabolism , Serine Proteinase Inhibitors/metabolism , Serpins/metabolism , Blood Proteins/chemistry , Blood Proteins/genetics , Blood Proteins/metabolism , Calcium/chemistry , Calcium/metabolism , Factor XIa/chemistry , Factor XIa/genetics , Factor XIa/metabolism , Factor Xa/chemistry , Factor Xa/genetics , Factor Xa/metabolism , Fibrinolytic Agents/chemistry , Heparin/chemistry , Humans , Lipids/chemistry , Lipids/genetics , Protein Binding/physiology , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/genetics , Serpins/chemistry , Serpins/genetics
11.
Arterioscler Thromb Vasc Biol ; 30(3): 388-92, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20139363

ABSTRACT

The dose-limiting issue with available anticoagulant therapies is bleeding. Is there an approach that could provide antithrombotic protection with reduced bleeding? One hypothesis is that targeting proteases upstream from the common pathway provides a reduction in thrombin sufficient to impede occlusive thrombosis yet allows enough thrombin generation to support hemostasis. The impairment of intrinsic coagulation by selective inhibition of factor XI (FXI) leaves the extrinsic and common pathways of coagulation intact, making FXI a drug target. This concept is supported by the observation that human deficiency in FXI results in a mild bleeding disorder compared with other coagulation factor deficiencies, and that elevated levels of FXI are a risk factor for thromboembolic disease. Moreover, FXI knockout mice have reduced thrombosis with little effect on hemostasis. The results from genetic models have been supported by studies using neutralizing antibodies, peptide inhibitors, and small-molecule inhibitors. These agents impede thrombosis without affecting bleeding time in a variety of experimental animals, including primates. Together, these data strongly support FXIa inhibition as a viable method to increase the ratio of benefit to risk in an antithrombotic drug.


Subject(s)
Anticoagulants/therapeutic use , Factor XIa/antagonists & inhibitors , Venous Thrombosis/drug therapy , Animals , Anticoagulants/adverse effects , Blood Coagulation/physiology , Disease Models, Animal , Factor XIa/genetics , Factor XIa/physiology , Hemorrhage/etiology , Hemorrhage/physiopathology , Hemostasis/physiology , Humans , Mice , Mice, Knockout , Risk Assessment , Venous Thrombosis/prevention & control
12.
J Biol Chem ; 283(27): 18655-64, 2008 Jul 04.
Article in English | MEDLINE | ID: mdl-18441012

ABSTRACT

Coagulation factor XI (FXI) is a covalent homodimer consisting of two identical subunits of 80 kDa linked by a disulfide bond formed by Cys-321 within the Apple 4 domain of each subunit. Because FXI(C321S) is a noncovalent dimer, residues within the interface between the two subunits must mediate its homodimeric structure. The crystal structure of FXI demonstrates formation of salt bridges between Lys-331 of one subunit and Glu-287 of the other subunit and hydrophobic interactions at the interface of the Apple 4 domains involving Ile-290, Leu-284, and Tyr-329. FXI(C321S), FXI(C321S,K331A), FXI(C321S,E287A), FXI(C321S,I290A), FXI(C321S,Y329A), FXI(C321S,L284A), FXI(C321S,K331R), and FXI(C321S,H343A) were expressed in HEK293 cells and characterized using size exclusion chromatography, analytical ultracentrifugation, electron microscopy, and functional assays. Whereas FXI(C321S) and FXI(C321S,H343A) existed in monomer/dimer equilibrium (K(d) approximately 40 nm), all other mutants were predominantly monomers with impaired dimer formation by analytical ultracentrifugation (K(d)=3-38 microm). When converted to the active enzyme, FXIa, all the monomeric mutants activated FIX similarly to wild-type dimeric FXIa. In contrast, these monomeric mutants could not be activated efficiently by FXIIa, thrombin, or autoactivation in the presence of dextran sulfate. We conclude that salt bridges formed between Lys-331 of one subunit and Glu-287 of the other together with hydrophobic interactions at the interface, involving residues Ile-290, Leu-284, and Tyr-329, are essential for homodimer formation. The dimeric structure of FXI is essential for normal proteolytic activation of FXI by FXIIa, thrombin, or FXIa either in solution or on an anionic surface but not for FIX activation by FXIa in solution.


Subject(s)
Factor XIIa/chemistry , Factor XI/chemistry , Factor XIa/chemistry , Thrombin/chemistry , Amino Acid Substitution , Crystallography, X-Ray , Dimerization , Disulfides/chemistry , Disulfides/metabolism , Factor XI/genetics , Factor XI/metabolism , Factor XIIa/genetics , Factor XIIa/metabolism , Factor XIa/genetics , Factor XIa/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Mutation, Missense , Protein Structure, Quaternary/genetics , Protein Structure, Tertiary/genetics , Thrombin/genetics , Thrombin/metabolism
14.
Biochemistry ; 47(5): 1326-35, 2008 Feb 05.
Article in English | MEDLINE | ID: mdl-18186617

ABSTRACT

In serine proteases, Gly193 (chymotrypsin numbering) is conserved with rare exception. Mutants of blood coagulation proteases have been reported with Glu, Ala, Arg or Val substitutions for Gly193. To further understand the role of Gly193 in protease activity, we replaced it with Ala or Val in coagulation factor XIa (FXIa). For comparison to the reported FXIa Glu193 mutant, we prepared FXIa with Asp (short side chain) or Lys (opposite charge) substitutions. Binding of p-aminobenzamidine (pAB) and diisopropylfluorphosphate (DFP) were impaired 1.6-36-fold and 35-478-fold, respectively, indicating distortion of, or altered accessibility to, the S1 and oxyanion-binding sites. Val or Asp substitutions caused the most impairment. Salt bridge formation between the amino terminus of the mature protease moiety at Ile16 and Asp194, essential for catalysis, was impaired 1.4-4-fold. Mutations reduced catalytic efficiency of tripeptide substrate hydrolysis 6-280-fold, with Val or Asp causing the most impairment. Further studies were directed toward macromolecular interactions with the FXIa mutants. kcat for factor IX activation was reduced 8-fold for Ala and 400-1100-fold for other mutants, while binding of the inhibitors antithrombin and amyloid beta-precursor protein Kunitz domain (APPI) was impaired 13-2300-fold and 22-27000-fold, respectively. The data indicate that beta-branching of the side chain of residue 193 is deleterious for interactions with pAB, DFP and amidolytic substrates, situations where no S2'-P2' interactions are involved. When an S2'-P2' interaction is involved (factor IX, antithrombin, APPI), beta-branching and increased side chain length are detrimental. Molecular models indicate that the mutants have impaired S2' binding sites and that beta-branching causes steric conflicts with the FXIa 140-loop, which could perturb the local tertiary structure of the protease domain. In conclusion, enzyme activity is impaired in FXIa when Gly193 is replaced by a non-Gly residue, and residues with side chains that branch at the beta-carbon have the greatest effect on catalysis and binding of substrates.


Subject(s)
Factor XIa/chemistry , Amino Acid Sequence , Amino Acid Substitution , Amyloid beta-Protein Precursor/pharmacology , Antithrombins/pharmacology , Benzamidines/chemistry , Carbamates/chemistry , Factor IX/metabolism , Factor XIa/antagonists & inhibitors , Factor XIa/genetics , Factor XIa/metabolism , Humans , Isoflurophate/pharmacology , Kinetics , Models, Molecular
15.
Biochemistry ; 46(50): 14450-60, 2007 Dec 18.
Article in English | MEDLINE | ID: mdl-18020374

ABSTRACT

The zymogen, factor XI, and the enzyme, factor XIa, interact specifically with functional receptors on the surface of activated platelets. These studies were initiated to identify the molecular subdomain within factor XIa that binds to activated platelets. Both factor XIa (Ki approximately 1.4 nM) and a chimeric factor XIa containing the Apple 3 domain of prekallikrein (Ki approximately 2.7 nM) competed with [125I]factor XIa for binding sites on activated platelets, suggesting that the factor XIa binding site for platelets is not located in the Apple 3 domain which mediates factor XI binding to platelets. The recombinant catalytic domain (Ile370-Val607) inhibited the binding of [125I]factor XIa to the platelets (Ki approximately 3.5 nM), whereas the recombinant factor XI heavy chain did not, demonstrating that the platelet binding site is located in the light chain of factor XIa. A conformationally constrained cyclic peptide (Cys527-Cys542) containing a high-affinity (KD approximately 86 nM) heparin-binding site within the catalytic domain of factor XIa also displaced [125I]factor XIa from the surface of activated platelets (Ki approximately 5.8 nM), whereas a scrambled peptide of identical composition was without effect, suggesting that the binding site in factor XIa that interacts with the platelet surface resides in the catalytic domain near the heparin binding site of factor XIa. These data support the conclusion that a conformational transition accompanies conversion of factor XI to factor XIa that conceals the Apple 3 domain factor XI (zymogen) platelet binding site and exposes the factor XIa (enzyme) platelet binding site within the catalytic domain possibly comprising residues Cys527-Cys542.


Subject(s)
Blood Platelets/metabolism , Factor XIa/chemistry , Factor XIa/metabolism , Amino Acid Sequence , Binding Sites , Catalysis , Cell Line , Cysteine/chemistry , Cysteine/genetics , Cysteine/metabolism , Enzyme-Linked Immunosorbent Assay , Factor XI/chemistry , Factor XI/genetics , Factor XI/metabolism , Factor XIIa/chemistry , Factor XIIa/genetics , Factor XIIa/metabolism , Factor XIa/genetics , Humans , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Prekallikrein/chemistry , Prekallikrein/genetics , Prekallikrein/metabolism , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Amino Acid
16.
Biochemistry ; 45(31): 9427-33, 2006 Aug 08.
Article in English | MEDLINE | ID: mdl-16878977

ABSTRACT

The autolysis loops (amino acids 143-154, chymotrypsinogen numbering) of plasma serine proteases play key roles in determining the specificity of protease inhibition by plasma serpins. We studied the importance of four basic residues (Arg-144, Lys-145, Arg-147, and Lys-149) in the autolysis loop of the coagulation protease factor XIa (fXIa) for inhibition by serpins. Recombinant fXIa mutants, in which these residues were replaced individually or in combination with alanine, were prepared. The proteases were compared to wild-type fXIa (fXIa-WT) with respect to their ability to activate factor IX in a plasma clotting assay, to hydrolyze the chromogenic substrate S2366, and to undergo inhibition by the C1-inhibitor (C1-INH), protein Z dependent protease inhibitor (ZPI), antithrombin (AT), and alpha(1)-protease inhibitor (alpha(1)-PI). All mutants exhibited normal activity in plasma and hydrolyzed S2366 with catalytic efficiencies similar to that of fXIa-WT. Inhibition of mutants by C1-INH was increased to varying degrees relative to that of fXIa-WT, with the mutant containing alanine replacements for all four basic residues (fXIa-144-149A) exhibiting an approximately 15-fold higher rate of inhibition. In contrast, the inhibition by ZPI was impaired 2-3-fold for single amino acid substitutions, and fXIa-144-149A was essentially resistant to inhibition by ZPI. Alanine substitution for Arg-147 impaired inhibition by AT approximately 7-fold; however, other substitutions did not affect it or slightly enhanced inhibition. Arg-147 was also required for inhibition by alpha(1)-PI. Cumulatively, the results demonstrate that basic amino acids in the autolysis loop of fXIa are important determinants of serpin specificity.


Subject(s)
Amino Acids, Basic/chemistry , Autolysis/genetics , Factor XIa/antagonists & inhibitors , Factor XIa/chemistry , Serpins/pharmacology , Amino Acid Sequence , Amino Acids, Basic/genetics , Antithrombins/pharmacology , Complement C1 Inhibitor Protein/pharmacology , Factor XIa/genetics , Humans , Molecular Sequence Data , Mutation , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
17.
Acta Crystallogr D Biol Crystallogr ; 61(Pt 10): 1418-25, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16204896

ABSTRACT

Activated factor XI (FXIa) is a key enzyme in the amplification phase of the blood-coagulation cascade. Thus, a selective FXIa inhibitor may have lesser bleeding liabilities and provide a safe alternative for antithrombosis therapy to available drugs on the market. In a previous report, the crystal structures of the catalytic domain of FXIa (rhFXI(370-607)) in complex with various ecotin mutants have been described. However, ecotin forms a matrix-like interaction with rhFXI(370-607) and is impossible to displace with small-molecule inhibitors; ecotin crystals are therefore not suitable for iterative structure-based ligand design. In addition, rhFXI(370-607) did not crystallize in the presence of small-molecule ligands. In order to obtain the crystal structure of rhFXI(370-607) with a weak small-molecule ligand, namely benzamidine, several rounds of surface-residue mutation were implemented to promote crystal formation of rhFXI(370-607). A quadruple mutant of rhFXI(370-607) (rhFXI(370-607)-S434A,T475A,C482S,K437A) readily crystallized in the presence of benzamidine. The benzamidine in the preformed crystals was easily exchanged with other FXIa small-molecule inhibitors. These crystals have facilitated the structure-based design of small-molecule FXIa inhibitors.


Subject(s)
Benzamidines/chemistry , Factor XI/antagonists & inhibitors , Factor XIa/chemistry , Factor XIa/genetics , Mutation , Binding Sites , Blood Coagulation Factors/chemistry , Catalysis , Catalytic Domain , Crystallography, X-Ray , DNA, Complementary/metabolism , Humans , Hydrogen Bonding , Inhibitory Concentration 50 , Ligands , Macromolecular Substances/chemistry , Models, Molecular , Mutagenesis , Mutagenesis, Site-Directed , Peptides/chemistry , Pichia/metabolism , Protein Binding , Protein Conformation , Recombinant Proteins/chemistry , Serine/chemistry
18.
Biochemistry ; 44(30): 10416-22, 2005 Aug 02.
Article in English | MEDLINE | ID: mdl-16042419

ABSTRACT

Factor XI, unlike other coagulation proteins, is a homodimer of two identical subunits linked by a single disulfide bond formed by Cys321. The present study was undertaken to understand the physiological significance of the dimeric nature of factor XI. We have expressed a mutant FXI/G326C in which the Gly326 residue of factor XI has been mutated to Cys326, reasoning that Cys321 would form an intrachain disulfide bond with Cys326 as in prekallikrein, a plasma protein that exists as a monomer even with 58% amino acid sequence identity and a domain structure very similar to factor XI. No free thiol could be detected in the expressed protein, and it migrated as a monomer on nonreduced SDS-PAGE. In physiological buffer, however, the protein was found to exist in a state of monomer-dimer equilibrium as assessed by gel-filtration chromatography and ultracentrifugation studies (K(d) approximately 36 nM). Functional studies revealed that FXI/G326C was indistinguishable from plasma factor XI in a plasma-clotting assay and in a factor IX activation assay both in the presence and absence of activated platelets even at concentrations at which less than 5% of the mutant exists as dimers. We conclude that, for optimal function in the presence of activated platelets, a preformed dimer of factor XI is not required.


Subject(s)
Factor IX/metabolism , Factor XIa/chemistry , Factor XIa/physiology , Amino Acid Substitution/genetics , Cell Line , Cysteine/genetics , Dextran Sulfate/pharmacology , Dimerization , Factor IX/chemistry , Factor XIIa/physiology , Factor XIa/genetics , Glycine/genetics , Humans , Hydrolysis , Oligopeptides/metabolism , Partial Thromboplastin Time/methods , Platelet Activation/genetics , Pyrrolidonecarboxylic Acid/analogs & derivatives , Pyrrolidonecarboxylic Acid/metabolism , Substrate Specificity/genetics , Thrombin/physiology
19.
J Biol Chem ; 279(28): 29485-92, 2004 Jul 09.
Article in English | MEDLINE | ID: mdl-15090552

ABSTRACT

In serine proteases, Gly(193) is highly conserved with few exceptions. A patient with inherited deficiency of the coagulation serine protease factor XI (FXI) was reported to be homozygous for a Gly(555) --> Glu substitution. Gly(555) in FXI corresponds to Gly(193) in chymotrypsin, which is the numbering system used subsequently. To investigate the abnormality in FXI(G193E), we expressed and purified recombinant FXIa(G193E), activated it to FXIa(G193E), and compared its activity to wild type-activated FXI (FXIa(WT)). FXIa(G193E) activated FIX with approximately 300-fold reduced k(cat) and similar K(m), and hydrolyzed synthetic substrate with approximately 10-fold reduced K(m) and modestly reduced k(cat). Binding of antithrombin and the amyloid beta-precursor protein Kunitz domain inhibitor (APPI) to FXIa(G193E) was impaired approximately 8000- and approximately 100000-fold, respectively. FXIa(G193E) inhibition by diisopropyl fluoro-phosphate was approximately 30-fold slower and affinity for p-aminobenzamidine (S1 site probe) was 6-fold weaker than for FXIa(WT). The rate of carbamylation of NH(2)-Ile(16), which forms a salt bridge with Asp(194) in active serine proteases, was 4-fold faster for FXIa(G193E). These data indicate that the unoccupied active site of FXIa(G193E) is incompletely formed, and the amide N of Glu(193) may not point toward the oxyanion hole. Inclusion of saturating amounts of p-aminobenzamidine resulted in comparable rates of carbamylation for FXIa(WT) and FXIa(G193E), suggesting that the occupied active site has near normal conformation. Thus, binding of small synthetic substrates or inhibitors provides sufficient energy to allow the amide N of Glu(193) to point correctly toward the oxyanion hole. Homology modeling also indicates that the inability of FXIa(G193E) to bind antithrombin/APPI or activate FIX is caused, in part, by impaired accessibility of the S2' site because of a steric clash with Glu(193). Such arguments will apply to other serine proteases with substitutions of Gly(193) with a non-glycine residue.


Subject(s)
Factor XIa/genetics , Factor XIa/metabolism , Glycine/metabolism , Point Mutation , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Animals , Chymotrypsin/chemistry , Chymotrypsin/genetics , Chymotrypsin/metabolism , Enzyme Activation , Enzyme Inhibitors/metabolism , Factor XIa/chemistry , Humans , Models, Molecular , Peptides/metabolism , Protein Binding , Protein Conformation , Serine Endopeptidases/chemistry
20.
Blood ; 97(10): 3117-22, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11342438

ABSTRACT

Human coagulation factor XI (FXI) is a plasma serine protease composed of 2 identical 80-kd polypeptides connected by a disulfide bond. This dimeric structure is unique among blood coagulation enzymes. The hypothesis was tested that dimeric conformation is required for normal FXI function by generating a monomeric version of FXI (FXI/PKA4) and comparing it to wild-type FXI in assays requiring factor IX activation by activated FXI (FXIa). FXI/PKA4 was made by replacing the FXI A4 domain with the A4 domain from prekallikrein (PK). A dimeric version of FXI/PKA4 (FXI/PKA4-Gly326) was prepared as a control. Activated FXI/PKA4 and FXI/PKA4-Gly326 activate factor IX with kinetic parameters similar to those of FXIa. In kaolin-triggered plasma clotting assays containing purified phospholipid, FXI/PKA4 and FXI/PKA4-Gly326 have coagulant activity similar to FXI. The surface of activated platelets is likely to be a physiologic site for reactions involving FXI/FXIa. In competition binding assays FXI/PKA4, FXI/PKA4-Gly326, and FXI have similar affinities for activated platelets (K(i) = 12-16 nM). In clotting assays in which phospholipid is replaced by activated platelets, the dimeric proteins FXI and FXI/PKA4-Gly326 promote coagulation similarly; however, monomeric FXI/PKA4 has greatly reduced activity. Western immunoblot analysis confirmed that activated monomeric FXI/PKA4 activates factor IX poorly in the presence of activated platelets. These findings demonstrate the importance of the dimeric state to FXI activity and suggest a novel model for factor IX activation in which FXIa binds to activated platelets by one chain of the dimer, while binding to factor IX through the other.


Subject(s)
Blood Platelets/chemistry , Dimerization , Factor IX/physiology , Factor XIa/chemistry , Factor XIa/physiology , Blood Coagulation , Blood Platelets/physiology , Blotting, Western , Cell Membrane/chemistry , Enzyme Activation , Factor IXa/physiology , Factor XI/physiology , Factor XIa/genetics , Humans , Kinetics , Platelet Activation , Prekallikrein/chemistry , Prekallikrein/genetics , Protein Conformation , Recombinant Fusion Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...