182 results on '"Stafford DW"'
Search Results
2. Structure and function of vitamin K epoxide reductase.
- Author
-
Tie JK, Stafford DW, Tie, Jian-Ke, and Stafford, Darrel W
- Abstract
Vitamin K epoxide reductase (VKOR) is an integral membrane protein that catalyzes the reduction of vitamin K 2,3-epoxide and vitamin K to vitamin K hydroquinone, a cofactor required for the gamma-glutamyl carboxylation reaction. VKOR is highly sensitive to inhibition by warfarin, the most commonly prescribed oral anticoagulant. Warfarin inhibition of VKOR decreases the concentration of reduced vitamin K, which reduces the rate of vitamin K-dependent carboxylation and leads to under-carboxylated, inactive vitamin K-dependent proteins. It is proposed that an active site disulfide needs to be reduced for the enzyme to be active. VKOR uses two sulfhydryl groups for the catalytic reaction and these two sulfhydryl groups are oxidized back to a disulfide bond during each catalytic cycle. The recent identification of the gene encoding VKOR allows us to study its structure and function relationship at the molecular level. The membrane topology model shows that VKOR spans the endoplasmic reticulum membrane three times with its amino-terminus residing in the lumen and the carboxyl-terminus residing in the cytoplasm. Both the active site (cysteines 132 and 135) and the proposed warfarin binding site (tyrosine 139) reside in the third transmembrane helix. VKOR is made at high levels in insect cells and is relatively easily purified. This should allow the determination of its three-dimensional structure. A detailed mechanism has been published and the purified enzyme should allow the testing of this mechanism. A major unanswered question is the physiological reductant of VKOR. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
3. Molecular cloning of a cDNA encoding canine factor IX
- Author
-
Evans, JP, Watzke, HH, Ware, JL, Stafford, DW, and High, KA
- Abstract
Factor IX (F.IX) is a vitamin K-dependent plasma protein, a deficiency of which results in hemophilia B. A canine model of hemophilia B exists; attempts to use this model for gene transfer experiments or characterization of the hemophilic defect require elucidation of normal canine F.IX structure. We report the isolation and characterization of the coding region for canine F.IX cDNA. Canine F.IX possesses 86% identity at the amino-acid level with human F.IX. The leader peptide, Gla domain, EGF domains, and the carboxy-terminal portion of the heavy chains show extensive sequence conservation between the canine and human. All Glu residues undergoing gamma-carboxylation in humans are conserved in canines. The complete coding sequence for canine F.IX has been determined, and the derived translation product has been analyzed. A similar approach should allow identification of the causative mutation in canine hemophilia B. Furthermore, this clone may prove a valuable resource in gene transfer experiments for this disease.
- Published
- 1989
- Full Text
- View/download PDF
4. Molecular defect in factor IXHilo, a hemophilia Bm variant: Arg----Gln at the carboxyterminal cleavage site of the activation peptide
- Author
-
Huang, MN, Kasper, CK, Roberts, HR, Stafford, DW, and High, KA
- Abstract
A genomic DNA library and the enzymatic DNA amplification technique were used to isolate human factor IX coding sequences of a hemophilia Bm variant, factor IXHilo. A point mutation that resulted in the substitution of a glutamine (CAG) for an arginine (CGG) at amino acid 180 was found in exon VI of the factor IX gene (G----A at nucleotide 20519). This mutation alters the carboxy terminal cleavage site for the activation peptide at Arg180-Val181. The arginine residue at the activation peptide cleavage site is conserved in mouse, canine, bovine, and human factor IX, suggesting that the arginine at amino acid 180 is important for normal cleavage. Sequencing of all of the coding regions of factor IXHilo revealed no other mutations. We have also shown that the point mutation in exon VI creates a new Dde I restriction site, which, in combination with the enzymatic DNA amplification technique, provides a quick, reliable, and sensitive method for carrier detection and antenatal diagnosis in affected kindreds. This is the first report of the molecular defect in a hemophilia Bm patient with a markedly prolonged ox brain prothrombin time.
- Published
- 1989
- Full Text
- View/download PDF
5. Factor IXAlabama: a point mutation in a clotting protein results in hemophilia B
- Author
-
Davis, LM, McGraw, RA, Ware, JL, Roberts, HR, and Stafford, DW
- Abstract
Factor IXAlabama is a variant factor IX molecule responsible for a clinically moderate form of hemophilia B. Twenty-five kilobases (kb) of the variant gene, including seven exons coding for the structural protein, were cloned and characterized. The restriction map and the arrangement of coding regions are identical to those of the normal gene. DNA sequence analysis of the coding regions revealed a single base-pair difference between the gene for factor IXAlabama and the normal factor IX gene. An adenine to guanine transition in the first nucleotide of exon d causes the substitution of a glycine codon (GGT) for the normal aspartic acid codon (GAT). This point mutation results in a single amino acid substitution at residue 47 of the zymogen and represents the genetic defect in factor IXAlabama.
- Published
- 1987
- Full Text
- View/download PDF
6. Mapping of monoclonal antibodies to human factor IX
- Author
-
Frazier, D, Smith, KJ, Cheung, WF, Ware, J, Lin, SW, Thompson, AR, Reisner, H, Bajaj, SP, and Stafford, DW
- Abstract
We used recombinant DNA techniques to map a panel of six monoclonal antibodies (MoAbs) to regions of the human factor IX molecule. A-2 maps to 17 amino acids at the amino terminus of the heavy chain of IXa; 2D5, an inhibitor of clotting, is defined to 36 amino acids of the first EGF- like domain of human factor IX. A-4, A-5, C10D, and FXC008 all map to a region of the heavy chain containing amino acids 180 through 310, suggesting an immunodominant site. FXC008 has been reported to interfere with binding of factor IXa to factor VIII:Ca.
- Published
- 1989
- Full Text
- View/download PDF
7. Genetic defect responsible for the dysfunctional protein: factor IXLong Beach
- Author
-
Ware, J, primary, Davis, L, additional, Frazier, D, additional, Bajaj, SP, additional, and Stafford, DW, additional
- Published
- 1988
- Full Text
- View/download PDF
8. Identification of a F.VIII epitope recognized by a human hemophilic inhibitor
- Author
-
Lubahn, BC, primary, Ware, J, additional, Stafford, DW, additional, and Reisner, HM, additional
- Published
- 1989
- Full Text
- View/download PDF
9. A genome-wide CRISPR-Cas9 knockout screen identifies FSP1 as the warfarin-resistant vitamin K reductase.
- Author
-
Jin DY, Chen X, Liu Y, Williams CM, Pedersen LC, Stafford DW, and Tie JK
- Subjects
- Humans, Anticoagulants pharmacology, CRISPR-Cas Systems, Ubiquinone pharmacology, Ubiquinone metabolism, Vitamin K metabolism, Vitamin K Epoxide Reductases genetics, Vitamin K Epoxide Reductases metabolism, NAD(P)H Dehydrogenase (Quinone) metabolism, Warfarin pharmacology, Apoptosis Regulatory Proteins genetics
- Abstract
Vitamin K is a vital micronutrient implicated in a variety of human diseases. Warfarin, a vitamin K antagonist, is the most commonly prescribed oral anticoagulant. Patients overdosed on warfarin can be rescued by administering high doses of vitamin K because of the existence of a warfarin-resistant vitamin K reductase. Despite the functional discovery of vitamin K reductase over eight decades ago, its identity remained elusive. Here, we report the identification of warfarin-resistant vitamin K reductase using a genome-wide CRISPR-Cas9 knockout screen with a vitamin K-dependent apoptotic reporter cell line. We find that ferroptosis suppressor protein 1 (FSP1), a ubiquinone oxidoreductase, is the enzyme responsible for vitamin K reduction in a warfarin-resistant manner, consistent with a recent discovery by Mishima et al. FSP1 inhibitor that inhibited ubiquinone reduction and thus triggered cancer cell ferroptosis, displays strong inhibition of vitamin K-dependent carboxylation. Intriguingly, dihydroorotate dehydrogenase, another ubiquinone-associated ferroptosis suppressor protein parallel to the function of FSP1, does not support vitamin K-dependent carboxylation. These findings provide new insights into selectively controlling the physiological and pathological processes involving electron transfers mediated by vitamin K and ubiquinone., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
10. Naturally occurring UBIAD1 mutations differentially affect menaquinone biosynthesis and vitamin K-dependent carboxylation.
- Author
-
Chen X, Furukawa N, Jin DY, Liu Y, Stafford DW, Williams CM, Suhara Y, and Tie JK
- Subjects
- Animals, Corneal Dystrophies, Hereditary, HEK293 Cells, Humans, Mice, Mutation, Vitamin K 2 metabolism, Dimethylallyltranstransferase genetics, Dimethylallyltranstransferase metabolism, Vitamin K genetics, Vitamin K metabolism
- Abstract
UbiA prenyltransferase domain-containing protein-1 (UBIAD1) is responsible for the biosynthesis of menaquinone-4 (MK-4), a cofactor for extrahepatic carboxylation of vitamin K-dependent (VKD) proteins. Genetic variations of UBIAD1 are mainly associated with Schnyder corneal dystrophy (SCD), a disease characterized by abnormal accumulation of cholesterol in the cornea. Results from in vitro studies demonstrate that SCD-associated UBIAD1 mutations are defective in MK-4 biosynthesis. However, SCD patients do not exhibit typical phenotypes associated with defects of MK-4 or VKD carboxylation. Here, we coupled UBIAD1's biosynthetic activity of MK-4 with VKD carboxylation in HEK293 cells that stably express a chimeric VKD reporter protein. The endogenous Ubiad1 gene in these cells was knocked out by CRISPR-Cas9-mediated genome editing. The effect of UBIAD1 mutations on MK-4 biosynthesis and VKD carboxylation was evaluated in Ubiad1-deficient reporter cells by determining the production of MK-4 or by measuring the efficiency of reporter-protein carboxylation. Our results show that the hot-spot mutation N102S has a moderate impact on MK-4 biosynthesis (retained ˜ 82% activity) but does not affect VKD carboxylation. However, the G186R mutation significantly affected both MK-4 biosynthesis and VKD carboxylation. Other mutations exhibit varying degrees of effects on MK-4 biosynthesis and VKD carboxylation. These results are consistent with in vivo results obtained from gene knock-in mice and SCD patients. Our findings suggest that UBIAD1's MK-4 biosynthetic activity does not directly correlate with the phenotypes of SCD patients. The established cell-based assays in this study provide a powerful tool for the functional studies of UBIAD1 in a cellular milieu., (© 2021 Federation of European Biochemical Societies.)
- Published
- 2022
- Full Text
- View/download PDF
11. The Function of extravascular coagulation factor IX in haemostasis.
- Author
-
Mann DM, Stafford KA, Poon MC, Matino D, and Stafford DW
- Subjects
- Animals, Half-Life, Hemorrhage prevention & control, Hemostasis, Humans, Mice, Factor IX genetics, Hemophilia B drug therapy
- Abstract
Introduction: The majority of clotting factor IX (FIX) resides extravascularly, in the subendothelial basement membrane, where it is important for haemostasis., Aim: We summarize preclinical studies demonstrating extravascular FIX and its role in haemostasis and discuss clinical observations supporting this. We compare the in vivo binding of BeneFIX
® and the extended half-life FIX, Alprolix® , to extravascular type IV collagen (Col4)., Methods: Three mouse models of haemophilia were used: the FIX knockout as the CRM- model and two knock-in mice, representing a CRM+ model of a commonly occurring patient mutation (FIXR333Q ) or a mutation that binds poorly to Col4 (FIXK5A ). The murine saphenous vein bleeding model was used to assess haemostatic competency. Clinical publications were reviewed for relevance to extravascular FIX., Results: CRM status affects recovery and prophylactic efficacy. Prophylactic protection decreases ~5X faster in CRM+ animals. Extravascular haemostasis can explain unexpected breakthrough bleeding in patients treated with some EHL-FIX therapeutics. In mice, both Alprolix® and BeneFIX® bind Col4 with similar affinities (Kd~20-40 nM) and show dose-dependent recoveries. As expected, the concentration of binding sites in the mouse calculated for Alprolix® (574 nM) was greater than for BeneFIX® (405 nM), due to Alprolix® binding to both Col4 and the endothelial cell neonatal Fc receptor., Conclusion: Preclinical and clinical results support the interpretation that FIX plays a role in haemostasis from its extravascular location. We believe that knowing the CRM status of haemophilia B patients is important for optimizing prophylactic dosing with less trial and error, thereby decreasing clinical morbidity., (© 2021 John Wiley & Sons Ltd.)- Published
- 2021
- Full Text
- View/download PDF
12. A novel vitamin K derived anticoagulant tolerant to genetic variations of vitamin K epoxide reductase.
- Author
-
Chen X, Liu Y, Furukawa N, Jin DY, Paul Savage G, Stafford DW, Suhara Y, Williams CM, and Tie JK
- Subjects
- Animals, Blood Coagulation, Vitamin K 1, Vitamin K Epoxide Reductases genetics, Warfarin, Anticoagulants, Vitamin K
- Abstract
Background: Vitamin K antagonists (VKAs), such as warfarin, have remained the cornerstone of oral anticoagulation therapy in the prevention and treatment of thromboembolism for more than half a century. They function by impairing the biosynthesis of vitamin K-dependent (VKD) clotting factors through the inhibition of vitamin K epoxide reductase (VKOR). The challenge of VKAs therapy is their narrow therapeutic index and highly variable dosing requirements, which are partially the result of genetic variations of VKOR., Objectives: The goal of this study was to search for an improved VKA that is tolerant to the genetic variations of its target enzyme., Methods: A series of vitamin K derivatives with benzyl and related side-chain substitutions at the 3-position of 1,4-naphthoquinone were synthesized. The role of these compounds in VKD carboxylation was evaluated by mammalian cell-based assays and conventional in vitro activity assays., Results: Our results showed that replacing the phytyl side-chain with a methylene cyclooctatetraene (COT) moiety at the 3-position of vitamin K
1 converted it from a substrate to an inhibitor for VKD carboxylation. Strikingly, this COT-vitamin K derivative displayed a similar inhibition potency in warfarin-resistant VKOR mutations whose warfarin resistance varied more than 400-fold. Further characterization of COT-vitamin K for the inhibition of VKD carboxylation suggested that this compound targets multiple enzymes in the vitamin K redox cycle. Importantly, the anticoagulation effect of COT-vitamin K can be rescued with high doses of vitamin K1 ., Conclusion: We discovered a vitamin K analogue that functions as a VKA and is tolerant to genetic variations in the target enzyme., (© 2020 International Society on Thrombosis and Haemostasis.)- Published
- 2021
- Full Text
- View/download PDF
13. γ-Glutamyl carboxylase mutations differentially affect the biological function of vitamin K-dependent proteins.
- Author
-
Hao Z, Jin DY, Chen X, Schurgers LJ, Stafford DW, and Tie JK
- Subjects
- Amino Acid Sequence, Base Sequence, Calcium-Binding Proteins genetics, Calcium-Binding Proteins metabolism, Carbon-Carbon Ligases chemistry, Carboxy-Lyases chemistry, Extracellular Matrix Proteins genetics, Extracellular Matrix Proteins metabolism, Genes, Reporter, Genetic Association Studies, Genetic Pleiotropy, HEK293 Cells, Hemorrhagic Disorders drug therapy, Hemorrhagic Disorders genetics, Humans, Mutation, Mutation, Missense, Osteocalcin genetics, Osteocalcin metabolism, Protein C genetics, Protein C metabolism, Protein Domains, Protein Interaction Mapping, Protein Isoforms genetics, Protein Isoforms metabolism, RNA Precursors metabolism, RNA Splicing, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Structure-Activity Relationship, Vitamin K physiology, Vitamin K therapeutic use, Matrix Gla Protein, Carbon-Carbon Ligases genetics, Carboxy-Lyases genetics, Protein Processing, Post-Translational genetics
- Abstract
γ-Glutamyl carboxylase (GGCX) is an integral membrane protein that catalyzes posttranslational carboxylation of a number of vitamin K-dependent (VKD) proteins involved in a wide variety of physiologic processes, including blood coagulation, vascular calcification, and bone metabolism. Naturally occurring GGCX mutations are associated with multiple distinct clinical phenotypes. However, the genotype-phenotype correlation of GGCX remains elusive. Here, we systematically examined the effect of all naturally occurring GGCX mutations on the carboxylation of 3 structure-function distinct VKD proteins in a cellular environment. GGCX mutations were transiently introduced into GGCX-deficient human embryonic kidney 293 cells stably expressing chimeric coagulation factor, matrix Gla protein (MGP), or osteocalcin as VKD reporter proteins, and then the carboxylation efficiency of these reporter proteins was evaluated. Our results show that GGCX mutations differentially affect the carboxylation of these reporter proteins and the efficiency of using vitamin K as a cofactor. Carboxylation of these reporter proteins by a C-terminal truncation mutation (R704X) implies that GGCX's C terminus plays a critical role in the binding of osteocalcin but not in the binding of coagulation factors and MGP. This has been confirmed by probing the protein-protein interaction between GGCX and its protein substrates in live cells using bimolecular fluorescence complementation and chemical cross-linking assays. Additionally, using a minigene splicing assay, we demonstrated that several GGCX missense mutations affect GGCX's pre-messenger RNA splicing rather than altering the corresponding amino acid residues. Results from this study interpreted the correlation of GGCX's genotype and its clinical phenotypes and clarified why vitamin K administration rectified bleeding disorders but not nonbleeding disorders., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
14. A cell-based high-throughput screen identifies drugs that cause bleeding disorders by off-targeting the vitamin K cycle.
- Author
-
Chen X, Li C, Jin DY, Ingram B, Hao Z, Bai X, Stafford DW, Hu K, and Tie JK
- Subjects
- 4-Hydroxycoumarins adverse effects, 4-Hydroxycoumarins isolation & purification, 4-Hydroxycoumarins pharmacology, Animals, Anticoagulants adverse effects, Blood Coagulation drug effects, Cell Culture Techniques methods, Drug Evaluation, Preclinical methods, HEK293 Cells, Hep G2 Cells, Humans, Indenes adverse effects, Indenes isolation & purification, Indenes pharmacology, Male, Metabolic Networks and Pathways drug effects, Mice, Mice, Inbred BALB C, Off-Label Use, Vitamin K adverse effects, Vitamin K antagonists & inhibitors, Vitamin K isolation & purification, Vitamin K pharmacology, Vitamin K Epoxide Reductases antagonists & inhibitors, Vitamin K Epoxide Reductases metabolism, Anticoagulants isolation & purification, Anticoagulants pharmacology, Hemorrhagic Disorders chemically induced, High-Throughput Screening Assays methods, Vitamin K metabolism
- Abstract
Drug-induced bleeding disorders contribute to substantial morbidity and mortality. Antithrombotic agents that cause unintended bleeding of obvious cause are relatively easy to control. However, the mechanisms of most drug-induced bleeding disorders are poorly understood, which makes intervention more difficult. As most bleeding disorders are associated with the dysfunction of coagulation factors, we adapted our recently established cell-based assay to identify drugs that affect the biosynthesis of active vitamin K-dependent (VKD) coagulation factors with possible adverse off-target results. The National Institutes of Health (NIH) Clinical Collection (NCC) library containing 727 drugs was screened, and 9 drugs were identified, including the most commonly prescribed anticoagulant warfarin. Bleeding complications associated with most of these drugs have been clinically reported, but the pathogenic mechanisms remain unclear. Further characterization of the 9 top-hit drugs on the inhibition of VKD carboxylation suggests that warfarin, lansoprazole, and nitazoxanide mainly target vitamin K epoxide reductase (VKOR), whereas idebenone, clofazimine, and AM404 mainly target vitamin K reductase (VKR) in vitamin K redox cycling. The other 3 drugs mainly affect vitamin K availability within the cells. The molecular mechanisms underlying the inactivation of VKOR and VKR by these drugs are clarified. Results from both cell-based and animal model studies suggest that the anticoagulation effect of drugs that target VKOR, but not VKR, can be rescued by the administration of vitamin K. These findings provide insights into the prevention and management of drug-induced bleeding disorders. The established cell-based, high-throughput screening approach provides a powerful tool for identifying new vitamin K antagonists that function as anticoagulants., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
15. Vitamin K-dependent carboxylation of coagulation factors: insights from a cell-based functional study.
- Author
-
Hao Z, Jin DY, Stafford DW, and Tie JK
- Subjects
- Animals, Factor IX genetics, Humans, Phenotype, Protein Processing, Post-Translational, Carbon-Carbon Ligases genetics, Carbon-Carbon Ligases metabolism, Vitamin K
- Abstract
Vitamin K-dependent carboxylation is a post-translational modification essential for the biological function of coagulation factors. Defects in carboxylation are mainly associated with bleeding disorders. With the discovery of new vitamin K-dependent proteins, the importance of carboxylation now encompasses vascular calcification, bone metabolism, and other important physiological processes. Our current knowledge of carboxylation, however, comes mainly from in vitro studies carried out under artificial conditions, which have a limited usefulness in understanding the carboxylation of vitamin K-dependent proteins in native conditions. Using a recently established mammalian cell-based assay, we studied the carboxylation of coagulation factors in a cellular environment. Our results show that the coagulation factor's propeptide controls substrate binding and product releasing during carboxylation, and the propeptide of factor IX appears to have the optimal affinity for efficient carboxylation. Additionally, non-conserved residues in the propeptide play an important role in carboxylation. A cell-based functional study of naturally occurring mutations in the propeptide successfully interpreted the clinical phenotype of warfarin's hypersensitivity during anticoagulation therapy in patients with these mutations. Unlike results obtained from in vitro studies, results from our cell-based study indicate that although the propeptide of osteocalcin cannot direct the carboxylation of the coagulation factor, it is required for the efficient carboxylation of osteocalcin. This suggests that the coagulation factors may have a different mechanism of carboxylation from osteocalcin. Together, results from this study provide insight into efficiently controlling one physiological process, such as coagulation without affecting the other, like bone metabolism., (Copyright© 2020 Ferrata Storti Foundation.)
- Published
- 2020
- Full Text
- View/download PDF
16. Dysfunctional endogenous FIX impairs prophylaxis in a mouse hemophilia B model.
- Author
-
Cooley B, Broze GJ Jr, Mann DM, Lin FC, Pedersen LG, and Stafford DW
- Subjects
- Animals, Collagen Type IV genetics, Collagen Type IV metabolism, Disease Models, Animal, Mice, Mice, Transgenic, Factor IX pharmacology, Hemophilia B blood, Hemophilia B drug therapy, Hemophilia B genetics, Recombinant Fusion Proteins pharmacology, Serum Albumin pharmacology
- Abstract
Factor IX (FIX) binds to collagen IV (Col4) in the subendothelial basement membrane. In hemophilia B, this FIX-Col4 interaction reduces the plasma recovery of infused FIX and plays a role in hemostasis. Studies examining the recovery of infused BeneFix (FIX
WT ) in null (cross-reactive material negative, CRM- ) hemophilia B mice suggest the concentration of Col4 readily available for binding FIX is ∼405 nM with a 95% confidence interval of 374 to 436 nM. Thus, the vascular cache of FIX bound to Col4 is several-fold the FIX level measured in plasma. In a mouse model of prophylactic therapy (testing hemostasis by saphenous vein bleeding 7 days after infusion of 150 IU/kg FIX), FIXWT and the increased half-life FIXs Alprolix (FIXFC ) and Idelvion (FIXAlb ) produce comparable hemostatic results in CRM- mice. In bleeding CRM- hemophilia B mice, the times to first clot at a saphenous vein injury site after the infusions of the FIX agents are significantly different, at FIXWT < FIXFC < FIXAlb Dysfunctional forms of FIX, however, circulate in the majority of patients with hemophilia B (CRM+ ). In the mouse prophylactic therapy model, none of the FIX products improves hemostasis in CRM+ mice expressing a dysfunctional FIX, FIXR333Q , that nevertheless competes with infused FIX for Col4 binding and potentially other processes involving FIX. The results in this mouse model of CRM+ hemophilia B demonstrate that the endogenous expression of a dysfunctional FIX can deleteriously affect the hemostatic response to prophylactic therapy., (© 2019 by The American Society of Hematology.)- Published
- 2019
- Full Text
- View/download PDF
17. Evaluation of oral anticoagulants with vitamin K epoxide reductase in its native milieu.
- Author
-
Chen X, Jin DY, Stafford DW, and Tie JK
- Subjects
- Administration, Oral, Anticoagulants administration & dosage, Cell Line, Drug Resistance, Enzyme Inhibitors administration & dosage, Humans, Phenindione administration & dosage, Phenindione analogs & derivatives, Phenindione pharmacology, Point Mutation, Vitamin K metabolism, Vitamin K Epoxide Reductases genetics, Vitamin K Epoxide Reductases metabolism, Warfarin administration & dosage, Warfarin pharmacology, Anticoagulants pharmacology, Enzyme Inhibitors pharmacology, Vitamin K Epoxide Reductases antagonists & inhibitors
- Abstract
Warfarin, acenocoumarol, phenprocoumon, and fluindione are commonly prescribed oral anticoagulants for the prevention and treatment of thromboembolic disorders. These anticoagulants function by impairing the biosynthesis of active vitamin K-dependent coagulation factors through the inhibition of vitamin K epoxide reductase (VKOR). Genetic variations in VKOR have been closely associated with the resistant phenotype of oral anticoagulation therapy. However, the relative efficacy of these anticoagulants, their mechanisms of action, and their resistance variations among naturally occurring VKOR mutations remain elusive. Here, we explored these questions using our recently established cell-based VKOR activity assay with the endogenous VKOR function ablated. Our results show that the efficacy of these anticoagulants on VKOR inactivation, from most to least, is: acenocoumarol > phenprocoumon > warfarin > fluindione. This is consistent with their effective clinical dosages for stable anticoagulation control. Cell-based functional studies of how each of the 27 naturally occurring VKOR mutations responds to these 4 oral anticoagulants indicate that phenprocoumon has the largest resistance variation (up to 199-fold), whereas the resistance of acenocoumarol varies the least (<14-fold). Cell-based kinetics studies show that fluindione appears to be a competitive inhibitor of VKOR, whereas warfarin is likely to be a mixed-type inhibitor of VKOR. The anticoagulation effect of these oral anticoagulants can be reversed by the administration of a high dose of vitamin K, apparently due to the existence of a different enzyme that can directly reduce vitamin K. These findings provide new insights into the selection of oral anticoagulants, their effective dosage management, and their mechanisms of anticoagulation., (© 2018 by The American Society of Hematology.)
- Published
- 2018
- Full Text
- View/download PDF
18. Warfarin and vitamin K epoxide reductase: a molecular accounting for observed inhibition.
- Author
-
Wu S, Chen X, Jin DY, Stafford DW, Pedersen LG, and Tie JK
- Subjects
- Amino Acid Motifs, Amino Acid Substitution, Animals, Binding Sites, COS Cells, Chlorocebus aethiops, Cysteine chemistry, HEK293 Cells, Humans, Hydrogen Bonding, Kinetics, Models, Molecular, Molecular Dynamics Simulation, Mutation, Missense, Point Mutation, Protein Binding, Protein Conformation, Tyrosine chemistry, Vitamin K Epoxide Reductases chemistry, Vitamin K Epoxide Reductases deficiency, Vitamin K Epoxide Reductases metabolism, Vitamin K Epoxide Reductases antagonists & inhibitors, Warfarin pharmacology
- Abstract
Vitamin K epoxide reductase (VKOR), an endoplasmic reticulum membrane protein, is the key enzyme for vitamin K-dependent carboxylation, a posttranslational modification that is essential for the biological functions of coagulation factors. VKOR is the target of the most widely prescribed oral anticoagulant, warfarin. However, the topological structure of VKOR and the mechanism of warfarin's inhibition of VKOR remain elusive. Additionally, it is not clear why warfarin-resistant VKOR mutations identified in patients significantly decrease warfarin's binding affinity, but have only a minor effect on vitamin K binding. Here, we used immunofluorescence confocal imaging of VKOR in live mammalian cells and PEGylation of VKOR's endogenous cytoplasmic-accessible cysteines in intact microsomes to probe the membrane topology of human VKOR. Our results show that the disputed loop sequence between the first and second transmembrane (TM) domain of VKOR is located in the cytoplasm, supporting a 3-TM topological structure of human VKOR. Using molecular dynamics (MD) simulations, a T-shaped stacking interaction between warfarin and tyrosine residue 139, within the proposed TY
139 A warfarin-binding motif, was observed. Furthermore, a reversible dynamic warfarin-binding pocket opening and conformational changes were observed when warfarin binds to VKOR. Several residues (Y25, A26, and Y139) were found essential for warfarin binding to VKOR by MD simulations, and these were confirmed by the functional study of VKOR and its mutants in their native milieu using a cell-based assay. Our findings provide new insights into the dynamics of the binding of warfarin to VKOR, as well as into warfarin's mechanism of anticoagulation., (© 2018 by The American Society of Hematology.)- Published
- 2018
- Full Text
- View/download PDF
19. Vitamin K epoxide reductase and its paralogous enzyme have different structures and functions.
- Author
-
Sinhadri BCS, Jin DY, Stafford DW, and Tie JK
- Subjects
- Amino Acid Sequence genetics, Cell Line, Cysteine chemistry, HEK293 Cells, Humans, Structure-Activity Relationship, Vitamin K Epoxide Reductases antagonists & inhibitors, Vitamin K Epoxide Reductases genetics, Warfarin pharmacology, Catalytic Domain genetics, Cell Membrane metabolism, Vitamin K metabolism, Vitamin K Epoxide Reductases metabolism
- Abstract
Vitamin K epoxide reductase (VKOR) is an essential enzyme for vitamin K-dependent carboxylation, while the physiological function of its paralogous enzyme VKOR-like (VKORL) is yet unknown. Although these two enzymes share approximately 50% protein sequence homology, the membrane topology of VKOR is still in debate. Here, we explored the differences in the membrane topology and disulfide-linked oligomerization of these two enzymes. Results from mutating the critical amino acid residues in the disputed transmembrane (TM) regions revealed that the second TM domain in the proposed 4-TM model of VKOR does not function as an authentic TM helix; supporting VKOR is a 3-TM protein, which is different from VKORL. Additionally, altering the loop sequence between the two conserved cysteine residues of VKORL affects its activity, supporting the notion that the conserved loop cysteines of VKORL are involved in its active site regeneration. However, a similar mutation in VKOR does not affect its enzymatic activity. Finally, our results show that although both VKOR and VKORL form disulfide-linked oligomers, the cysteine residues involved in the oligomerization appear to be different. Overall, the structural and functional differences between VKOR and VKORL shown here indicate that VKORL might have a different physiological function other than recycling vitamin K.
- Published
- 2017
- Full Text
- View/download PDF
20. Molecular basis of the first reported clinical case of congenital combined deficiency of coagulation factors.
- Author
-
Jin DY, Ingram BO, Stafford DW, and Tie JK
- Subjects
- Blood Coagulation Factors metabolism, CRISPR-Cas Systems, Carbon-Carbon Ligases genetics, Dose-Response Relationship, Drug, Gene Knockout Techniques, Genes, Reporter, HEK293 Cells, Hemorrhagic Disorders blood, Humans, Introns genetics, Protein Binding, Protein Domains, Protein Processing, Post-Translational, Protein Stability, RNA Splicing genetics, Vitamin K administration & dosage, Vitamin K pharmacology, Carbon-Carbon Ligases deficiency, Frameshift Mutation, Hemorrhagic Disorders genetics, Sequence Deletion
- Published
- 2017
- Full Text
- View/download PDF
21. Functional Study of the Vitamin K Cycle Enzymes in Live Cells.
- Author
-
Tie JK and Stafford DW
- Subjects
- Animals, Enzyme-Linked Immunosorbent Assay methods, Humans, Protein Processing, Post-Translational genetics, Vitamin K antagonists & inhibitors, Vitamin K metabolism, Vitamin K 1 analogs & derivatives, Vitamin K 1 chemistry, Enzyme Assays methods, NAD(P)H Dehydrogenase (Quinone) chemistry, Vitamin K chemistry, Vitamin K Epoxide Reductases chemistry
- Abstract
Vitamin K-dependent carboxylation, an essential posttranslational modification catalyzed by gamma-glutamyl carboxylase, is required for the biological functions of proteins that control blood coagulation, vascular calcification, bone metabolism, and other important physiological processes. Concomitant with carboxylation, reduced vitamin K (KH
2 ) is oxidized to vitamin K epoxide (KO). KO must be recycled back to KH2 by the enzymes vitamin K epoxide reductase and vitamin K reductase in a pathway known as the vitamin K cycle. Our current knowledge about the enzymes of the vitamin K cycle is mainly based on in vitro studies of each individual enzymes under artificial conditions, which are of limited usefulness in understanding how the complex carboxylation process is carried out in the physiological environment. In this chapter, we review the current in vitro activity assays for vitamin K cycle enzymes. We describe the rationale, establishment, and application of cell-based assays for the functional study of these enzymes in the native cellular milieu. In these cell-based assays, different vitamin K-dependent proteins were designed and stably expressed in mammalian cells as reporter proteins to accommodate the readily used enzyme-linked immunosorbent assay for carboxylation efficiency evaluation. Additionally, recently emerged genome-editing techniques TALENs and CRISPR-Cas9 were used to knock out the endogenous enzymes in the reporter cell lines to eliminate the background. These cell-based assays are easy to scale up for high-throughput screening of inhibitors of vitamin K cycle enzymes and have been successfully used to clarify the genotypes and their clinical phenotypes of enzymes of the vitamin K cycle., (© 2017 Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
22. Splice-Site Mutation of Exon 3 Deletion in the Gamma-Glutamyl Carboxylase Gene Causes Inactivation of the Enzyme.
- Author
-
Jin DY, Vermeer C, Stafford DW, and Tie JK
- Subjects
- Humans, Carbon-Carbon Ligases genetics, Exons, Mutation, RNA Splicing
- Abstract
Competing Interests: Dr. Vermeer received salary from VitaK. The other authors state no conflict of interest.
- Published
- 2016
- Full Text
- View/download PDF
23. Extravascular FIX and coagulation.
- Author
-
Stafford DW
- Abstract
This review summarizes the evidence that collagen IV binding is physiologically important, and that the extravascular compartment of FIX is composed of type IV collagen. As we have previously demonstrated, 7 days post-infusion, FIX
WT (BeneFIX) is able to control bleeding as well as the same dosage of Alprolix in hemophilia B mice, tested using the saphenous vein bleeding model (Alprolix is a chimeric FIX molecule joined at its C terminus to a Fc domain). Furthermore, we have shown that in hemophilia B mice, doses of BeneFIX or Alprolix (up to a dose of 150 IU/kg) have increased bleeding-control effectiveness in proportion to the dose up to a certain limit: higher doses are no more effective than the 150 IU/kg dose. These studies suggest that in hemophilia B mice, tested using the saphenous vein bleeding model, three things are true: first, extravascular FIX is at least as important for coagulation as is circulating FIX; second, measuring circulating levels of FIX may not be the best criterion for designing new "longer lasting" FIX molecules; and third, trough levels are less diagnostic for FIX therapy than they are for FVIII therapy.- Published
- 2016
- Full Text
- View/download PDF
24. Prophylactic efficacy of BeneFIX vs Alprolix in hemophilia B mice.
- Author
-
Cooley B, Funkhouser W, Monroe D, Ezzell A, Mann DM, Lin FC, Monahan PE, and Stafford DW
- Subjects
- Animals, Disease Models, Animal, Dose-Response Relationship, Drug, Mice, Factor IX pharmacokinetics, Factor IX pharmacology, Hemophilia B blood, Hemophilia B drug therapy, Hemorrhage blood, Hemorrhage prevention & control, Immunoglobulin Fc Fragments pharmacology, Recombinant Fusion Proteins pharmacokinetics, Recombinant Fusion Proteins pharmacology
- Abstract
FIX binds tightly to collagen IV. Furthermore, a FIX mutant, FIXK5R, which binds better than wild-type FIX to collagen IV, provides better hemostasis than wild-type FIX, long after both are undetectable in the plasma. There is also credible evidence of extravascular FIX. Here, we use the saphenous vein bleeding model to compare the efficacy of recombinant FIXFc (Alprolix) and wild-type FIX (BeneFIX) in hemophilia B mice 7 days postinfusion. Although the terminal half-life of Alprolix is significantly longer than that of BeneFIX, at equal doses Alprolix is not better at controlling bleeding 7 days postinfusion, presumably because of the extravascular FIX. Both BeneFIX and Alprolix exhibit a linear response in clotting efficacy up to 150 IU/kg, where they appear to saturate an extravascular compartment, because there is no additional prophylactic benefit from higher doses. A robust pool of extravascular FIX is clearly observed surrounding blood vessels, localized to the same region as collagen IV, in 2 representative human tissues: liver and skeletal muscle. We see no increased risk for thrombosis at 250 IU/kg FIX at 6 hours postinfusion. In summary, 7 days postinfusion into hemophilia B mice, BeneFIX and Alprolix are hemostatically indistinguishable despite the latter's increased half-life. We predict that doses of FIX ∼3 times higher than the currently recommended 40 to 50 IU/kg will, because of FIX's large extravascular compartment, efficiently prolong prophylactic hemostasis without thrombotic risk., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
25. Characterization of vitamin K-dependent carboxylase mutations that cause bleeding and nonbleeding disorders.
- Author
-
Tie JK, Carneiro JD, Jin DY, Martinhago CD, Vermeer C, and Stafford DW
- Subjects
- Abnormalities, Multiple genetics, Amino Acid Sequence, Base Sequence, Blood Coagulation Tests, CRISPR-Cas Systems, Calcinosis genetics, Calcium-Binding Proteins genetics, Cartilage Diseases genetics, DNA Mutational Analysis, Extracellular Matrix Proteins genetics, Female, Genes, Reporter, Genetic Association Studies, Genotype, HEK293 Cells, Hand Deformities, Congenital genetics, Hemorrhage, Humans, Infant, Infant, Newborn, Molecular Sequence Data, Phenotype, Pulmonary Valve Stenosis genetics, Sequence Homology, Amino Acid, Sequence Homology, Nucleic Acid, Matrix Gla Protein, Carbon-Carbon Ligases genetics, Mutation, Vitamin K Deficiency Bleeding genetics
- Abstract
Vitamin K-dependent coagulation factors deficiency is a bleeding disorder mainly associated with mutations in γ-glutamyl carboxylase (GGCX) that often has fatal outcomes. Some patients with nonbleeding syndromes linked to GGCX mutations, however, show no coagulation abnormalities. The correlation between GGCX genotypes and their clinical phenotypes has been previously unknown. Here we report the identification and characterization of novel GGCX mutations in a patient with both severe cerebral bleeding disorder and comorbid Keutel syndrome, a nonbleeding malady caused by functional defects of matrix γ-carboxyglutamate protein (MGP). To characterize GGCX mutants in a cellular milieu, we established a cell-based assay by stably expressing 2 reporter proteins (a chimeric coagulation factor and MGP) in HEK293 cells. The endogenous GGCX gene in these cells was knocked out by CRISPR-Cas9-mediated genome editing. Our results show that, compared with wild-type GGCX, the patient's GGCX D153G mutant significantly decreased coagulation factor carboxylation and abolished MGP carboxylation at the physiological concentration of vitamin K. Higher vitamin K concentrations can restore up to 60% of coagulation factor carboxylation but do not ameliorate MGP carboxylation. These results are consistent with the clinical results obtained from the patient treated with vitamin K, suggesting that the D153G alteration in GGCX is the causative mutation for both the bleeding and nonbleeding disorders in our patient. These findings provide the first evidence of a GGCX mutation resulting in 2 distinct clinical phenotypes; the established cell-based assay provides a powerful tool for studying the clinical consequences of naturally occurring GGCX mutations in vivo., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
26. Structural and functional insights into enzymes of the vitamin K cycle.
- Author
-
Tie JK and Stafford DW
- Subjects
- Amino Acid Sequence, Animals, Carbon-Carbon Ligases chemistry, Carbon-Carbon Ligases genetics, Gene Expression Regulation, Enzymologic, Genotype, Humans, Models, Molecular, Molecular Sequence Data, NAD(P)H Dehydrogenase (Quinone) genetics, Phenotype, Protein Conformation, Structure-Activity Relationship, Vitamin K Epoxide Reductases chemistry, Vitamin K Epoxide Reductases genetics, Blood Coagulation, Carbon-Carbon Ligases metabolism, NAD(P)H Dehydrogenase (Quinone) metabolism, Vitamin K metabolism, Vitamin K Epoxide Reductases metabolism
- Abstract
Vitamin K-dependent proteins require carboxylation of certain glutamates for their biological functions. The enzymes involved in the vitamin K-dependent carboxylation include: gamma-glutamyl carboxylase (GGCX), vitamin K epoxide reductase (VKOR) and an as-yet-unidentified vitamin K reductase (VKR). Due to the hydrophobicity of vitamin K, these enzymes are likely to be integral membrane proteins that reside in the endoplasmic reticulum. Therefore, structure-function studies on these enzymes have been challenging, and some of the results are notably controversial. Patients with naturally occurring mutations in these enzymes, who mainly exhibit bleeding disorders or are resistant to oral anticoagulant treatment, provide valuable information for the functional study of the vitamin K cycle enzymes. In this review, we discuss: (i) the discovery of the enzymatic activities and gene identifications of the vitamin K cycle enzymes; (ii) the identification of their functionally important regions and their active site residues; (iii) the membrane topology studies of GGCX and VKOR; and (iv) the controversial issues regarding the structure and function studies of these enzymes, particularly, the membrane topology, the role of the conserved cysteines and the mechanism of active site regeneration of VKOR. We also discuss the possibility that a paralogous protein of VKOR, VKOR-like 1 (VKORL1), is involved in the vitamin K cycle, and the importance of and possible approaches for identifying the unknown VKR. Overall, we describe the accomplishments and the remaining questions in regard to the structure and function studies of the enzymes in the vitamin K cycle., Competing Interests: of Conflict of Interests The authors hold a patent on the use of VKOR for producing vitamin K-dependent proteins licensed to Emergent., (© 2015 International Society on Thrombosis and Haemostasis.)
- Published
- 2016
- Full Text
- View/download PDF
27. Employing a gain-of-function factor IX variant R338L to advance the efficacy and safety of hemophilia B human gene therapy: preclinical evaluation supporting an ongoing adeno-associated virus clinical trial.
- Author
-
Monahan PE, Sun J, Gui T, Hu G, Hannah WB, Wichlan DG, Wu Z, Grieger JC, Li C, Suwanmanee T, Stafford DW, Booth CJ, Samulski JJ, Kafri T, McPhee SW, and Samulski RJ
- Subjects
- Animals, Antibodies, Neutralizing analysis, Capsid chemistry, Capsid immunology, Clinical Trials as Topic, Dependovirus immunology, Disease Models, Animal, Drug Evaluation, Preclinical, Factor IX metabolism, Factor IX pharmacokinetics, Gene Expression, Genetic Engineering, Genetic Vectors administration & dosage, Genetic Vectors chemistry, Hemophilia B blood, Hemophilia B genetics, Hemophilia B physiopathology, Hemorrhage blood, Hemorrhage genetics, Hemorrhage physiopathology, Humans, Liver immunology, Liver virology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Recombinant Proteins genetics, Recombinant Proteins metabolism, Recombinant Proteins pharmacokinetics, Tail, Tissue Distribution, Virion genetics, Dependovirus genetics, Factor IX genetics, Genetic Therapy methods, Genetic Vectors pharmacokinetics, Hemophilia B therapy, Hemorrhage prevention & control
- Abstract
Vector capsid dose-dependent inflammation of transduced liver has limited the ability of adeno-associated virus (AAV) factor IX (FIX) gene therapy vectors to reliably convert severe to mild hemophilia B in human clinical trials. These trials also identified the need to understand AAV neutralizing antibodies and empty AAV capsids regarding their impact on clinical success. To address these safety concerns, we have used a scalable manufacturing process to produce GMP-grade AAV8 expressing the FIXR338L gain-of-function variant with minimal (<10%) empty capsid and have performed comprehensive dose-response, biodistribution, and safety evaluations in clinically relevant hemophilia models. The scAAV8.FIXR338L vector produced greater than 6-fold increased FIX specific activity compared with wild-type FIX and demonstrated linear dose responses from doses that produced 2-500% FIX activity, associated with dose-dependent hemostasis in a tail transection bleeding challenge. More importantly, using a bleeding model that closely mimics the clinical morbidity of hemophilic arthropathy, mice that received the scAAV8.FIXR338L vector developed minimal histopathological findings of synovitis after hemarthrosis, when compared with mice that received identical doses of wild-type FIX vector. Hemostatically normal mice (n=20) and hemophilic mice (n=88) developed no FIX antibodies after peripheral intravenous vector delivery. No CD8(+) T cell liver infiltrates were observed, despite the marked tropism of scAAV8.FIXR338L for the liver in a comprehensive biodistribution evaluation (n=60 animals). With respect to the role of empty capsids, we demonstrated that in vivo FIXR338L expression was not influenced by the presence of empty AAV particles, either in the presence or absence of various titers of AAV8-neutralizing antibodies. Necropsy of FIX(-/-) mice 8-10 months after vector delivery revealed no microvascular or macrovascular thrombosis in mice expressing FIXR338L (plasma FIX activity, 100-500%). These preclinical studies demonstrate a safety:efficacy profile supporting an ongoing phase 1/2 human clinical trial of the scAAV8.FIXR338L vector (designated BAX335).
- Published
- 2015
- Full Text
- View/download PDF
28. Conserved loop cysteines of vitamin K epoxide reductase complex subunit 1-like 1 (VKORC1L1) are involved in its active site regeneration.
- Author
-
Tie JK, Jin DY, and Stafford DW
- Subjects
- Amino Acid Sequence, Cell Membrane metabolism, Disulfides chemistry, HEK293 Cells, Humans, Molecular Sequence Data, Oxidation-Reduction, Structure-Activity Relationship, Vitamin K analogs & derivatives, Vitamin K metabolism, Catalytic Domain, Conserved Sequence, Vitamin K Epoxide Reductases chemistry, Vitamin K Epoxide Reductases metabolism
- Abstract
Vitamin K epoxide reductase complex subunit 1 (VKORC1) reduces vitamin K epoxide in the vitamin K cycle for post-translational modification of proteins that are involved in a variety of biological functions. However, the physiological function of VKORC1-like 1 (VKORC1L1), a paralogous enzyme sharing about 50% protein identity with VKORC1, is unknown. Here we determined the structural and functional differences of these two enzymes using fluorescence protease protection (FPP) assay and an in vivo cell-based activity assay. We show that in vivo VKORC1L1 reduces vitamin K epoxide to support vitamin K-dependent carboxylation as efficiently as does VKORC1. However, FPP assays show that unlike VKORC1, VKORC1L1 is a four-transmembrane domain protein with both its termini located in the cytoplasm. Moreover, the conserved loop cysteines, which are not required for VKORC1 activity, are essential for VKORC1L1's active site regeneration. Results from domain exchanges between VKORC1L1 and VKORC1 suggest that it is VKORC1L1's overall structure that uniquely allows for active site regeneration by the conserved loop cysteines. Intermediate disulfide trapping results confirmed an intra-molecular electron transfer pathway for VKORC1L1's active site reduction. Our results allow us to propose a concerted action of the four conserved cysteines of VKORC1L1 for active site regeneration; the second loop cysteine, Cys-58, attacks the active site disulfide, forming an intermediate disulfide with Cys-139; the first loop cysteine, Cys-50, attacks the intermediate disulfide resulting in active site reduction. The different membrane topologies and reaction mechanisms between VKORC1L1 and VKORC1 suggest that these two proteins might have different physiological functions.
- Published
- 2014
- Full Text
- View/download PDF
29. FVIIa as used pharmacologically is not TF dependent in hemophilia B mice.
- Author
-
Feng D, Whinna H, Monroe D, and Stafford DW
- Subjects
- Animals, Binding Sites, Hemophilia B metabolism, Humans, Mice, Mice, Inbred C57BL, Models, Molecular, Saphenous Vein drug effects, Saphenous Vein pathology, Factor IX pharmacology, Factor VIIa pharmacology, Hemophilia B drug therapy, Hemorrhage prevention & control, Hemostasis drug effects, Thromboplastin metabolism
- Abstract
Activated factor VII is approved for treating hemophilia patients with autoantibodies to their factor IX or FVIII; however, its mechanism of action remains controversial. Some studies suggest that FVIIa requires tissue factor (TF) for function and that the reason for the high dose requirement is that it must compete with endogenous FVII for tissue factor. Others suggest that FVIIa binds platelets where it activates FX directly; the high concentration required would result from FVIIa's weak affinity for phospholipids. We address this question by infusing a chimera of mouse FIX (Gla and EGF1) with FVIIa (EGF2 and catalytic domain) into hemophilia B mice. This mutant has no TF-dependent activity because it cannot functionally bind TF at physiologically relevant concentrations. In vivo, this mutant is as effective as mouse FVIIa in controlling bleeding in hemophilia B mice. Our results suggest that the hemostatic effect of pharmacologic doses of FVIIa is TF independent.
- Published
- 2014
- Full Text
- View/download PDF
30. A conformational investigation of propeptide binding to the integral membrane protein γ-glutamyl carboxylase using nanodisc hydrogen exchange mass spectrometry.
- Author
-
Parker CH, Morgan CR, Rand KD, Engen JR, Jorgenson JW, and Stafford DW
- Subjects
- Humans, Peptides, Protein Binding, Protein Conformation, Carbon-Carbon Ligases chemistry, Carbon-Carbon Ligases metabolism, Hydrogen chemistry, Mass Spectrometry methods
- Abstract
Gamma (γ)-glutamyl carboxylase (GGCX) is an integral membrane protein responsible for the post-translational catalytic conversion of select glutamic acid (Glu) residues to γ-carboxy glutamic acid (Gla) in vitamin K-dependent (VKD) proteins. Understanding the mechanism of carboxylation and the role of GGCX in the vitamin K cycle is of biological interest in the development of therapeutics for blood coagulation disorders. Historically, biophysical investigations and structural characterizations of GGCX have been limited due to complexities involving the availability of an appropriate model membrane system. In previous work, a hydrogen exchange mass spectrometry (HX MS) platform was developed to study the structural configuration of GGCX in a near-native nanodisc phospholipid environment. Here we have applied the nanodisc-HX MS approach to characterize specific domains of GGCX that exhibit structural rearrangements upon binding the high-affinity consensus propeptide (pCon; AVFLSREQANQVLQRRRR). pCon binding was shown to be specific for monomeric GGCX-nanodiscs and promoted enhanced structural stability to the nanodisc-integrated complex while maintaining catalytic activity in the presence of carboxylation co-substrates. Noteworthy modifications in HX of GGCX were prominently observed in GGCX peptides 491-507 and 395-401 upon pCon association, consistent with regions previously identified as sites for propeptide and glutamate binding. Several additional protein regions exhibited minor gains in solvent protection upon propeptide incorporation, providing evidence for a structural reorientation of the GGCX complex in association with VKD carboxylation. The results herein demonstrate that nanodisc-HX MS can be utilized to study molecular interactions of membrane-bound enzymes in the absence of a complete three-dimensional structure and to map dynamic rearrangements induced upon ligand binding.
- Published
- 2014
- Full Text
- View/download PDF
31. Membrane topology for human vitamin K epoxide reductase.
- Author
-
Wu S, Tie JK, Stafford DW, and Pedersen LG
- Subjects
- Humans, Molecular Dynamics Simulation, Vitamin K Epoxide Reductases chemistry, Cell Membrane metabolism, Vitamin K Epoxide Reductases metabolism
- Published
- 2014
- Full Text
- View/download PDF
32. Evidence of clinically significant extravascular stores of factor IX.
- Author
-
Feng D, Stafford KA, Broze GJ, and Stafford DW
- Subjects
- Blood Coagulation Tests, Humans, Factor IX metabolism
- Published
- 2013
- Full Text
- View/download PDF
33. Assessment of the contribution of NAD(P)H-dependent quinone oxidoreductase 1 (NQO1) to the reduction of vitamin K in wild-type and NQO1-deficient mice.
- Author
-
Ingram BO, Turbyfill JL, Bledsoe PJ, Jaiswal AK, and Stafford DW
- Subjects
- Animals, Anticoagulants poisoning, Carbon-Carbon Ligases metabolism, Hemostasis, Kinetics, Male, Mice, Mice, Knockout, Microsomes, Liver enzymology, NAD(P)H Dehydrogenase (Quinone) genetics, Oxidation-Reduction, Warfarin poisoning, NAD(P)H Dehydrogenase (Quinone) metabolism, Vitamin K 2 metabolism
- Abstract
NQO1 [NAD(P)H quinone oxidoreductase 1; also known as DT-diaphorase] is a cytosolic enzyme that catalyses the two-electron reduction of various quinones including vitamin K. The enzyme may play a role in vitamin K metabolism by reducing vitamin K to vitamin K hydroquinone for utilization in the post-translational γ-glutamyl carboxylation reactions required by several proteins involved in blood coagulation. The aim of the present study was to assess the contribution of NQO1 to vitamin K reduction and haemostasis in an in vivo model. We examined the contribution of NQO1 to haemostasis by examining survival rates in mice poisoned with the anticoagulant warfarin. Supraphysiological amounts of vitamin K sufficiently reversed the effects of warfarin in both wild-type and NQO1-deficient mice. Additionally, vitamin K reductase activities distinct from VKOR (vitamin K epoxide reductase) and NQO1 were measured in vitro from both wild-type and NQO1-defecient mice. The results of the present study suggest that NQO1 does not play a major role in the production of vitamin K hydroquinone and supports the existence of multiple vitamin K reduction pathways. The properties of a NAD(P)H-dependent vitamin K reductase different from NQO1 are described.
- Published
- 2013
- Full Text
- View/download PDF
34. Evaluation of warfarin resistance using transcription activator-like effector nucleases-mediated vitamin K epoxide reductase knockout HEK293 cells.
- Author
-
Tie JK, Jin DY, Tie K, and Stafford DW
- Subjects
- Anticoagulants chemistry, Dithiothreitol chemistry, Gene Knockout Techniques, HEK293 Cells, Humans, Mutation, Oxygen chemistry, Phenotype, Polymorphism, Single Nucleotide, Transcription, Genetic, Vitamin K metabolism, Vitamin K Epoxide Reductases genetics, Drug Resistance, Vitamin K Epoxide Reductases metabolism, Warfarin chemistry
- Abstract
Background: Single nucleotide polymorphisms in the vitamin K epoxide reductase (VKOR) gene have been successfully used for warfarin dosage prediction. However, warfarin resistance studies of naturally occurring VKOR mutants do not correlate with their clinical phenotype. This discrepancy presumably arises because the in vitro VKOR activity assay is performed under artificial conditions using the non-physiological reductant dithiothreitol., Objectives: The aim of this study is to establish an in vivo VKOR activity assay in mammalian cells (HEK293) where VKOR functions in its native milieu without interference from endogenous enzymes., Methods: Endogenous VKOR activity in HEK293 cells was knocked out by transcription activator-like effector nucleases (TALENs)-mediated genome editing., Results and Conclusions: Knockout of VKOR in HEK293 cells significantly decreased vitamin K-dependent carboxylation with vitamin K epoxide (KO) as substrate. However, the paralog of VKOR, VKORC1L1, also exhibits substantial ability to convert KO to vitamin K for carboxylation. Using both VKOR and VKORC1L1 knockout cells, we examined the enzymatic activity and warfarin resistance of 10 naturally occurring VKOR mutants that were reported previously to have no activity in an in vitro assay. All 10 mutants are fully active; five have increased warfarin resistance, with the order being W59R>L128R≈W59L>N77S≈S52L. Except for the L128R mutant, this order is consistent with the clinical anticoagulant dosages. The other five VKOR mutants do not change VKOR's warfarin sensitivity, suggesting that factors other than VKOR play important roles. In addition, we confirmed that the conserved loop cysteines in VKOR are not required for active site regeneration after each cycle of oxidation., (© 2013 International Society on Thrombosis and Haemostasis.)
- Published
- 2013
- Full Text
- View/download PDF
35. Human vitamin K epoxide reductase and its bacterial homologue have different membrane topologies and reaction mechanisms.
- Author
-
Tie JK, Jin DY, and Stafford DW
- Subjects
- Bacterial Proteins genetics, Bacterial Proteins metabolism, Cell Membrane genetics, Crystallography, X-Ray, Cytoplasm genetics, Endoplasmic Reticulum genetics, HEK293 Cells, Humans, Mixed Function Oxygenases genetics, Mixed Function Oxygenases metabolism, Structural Homology, Protein, Synechococcus, Vitamin K Epoxide Reductases, Bacterial Proteins chemistry, Cell Membrane enzymology, Cytoplasm enzymology, Endoplasmic Reticulum enzymology, Mixed Function Oxygenases chemistry, Models, Molecular
- Abstract
Vitamin K epoxide reductase (VKOR) is essential for the production of reduced vitamin K that is required for modification of vitamin K-dependent proteins. Three- and four-transmembrane domain (TMD) topology models have been proposed for VKOR. They are based on in vitro glycosylation mapping of the human enzyme and the crystal structure of a bacterial (Synechococcus) homologue, respectively. These two models place the functionally disputed conserved loop cysteines, Cys-43 and Cys-51, on different sides of the endoplasmic reticulum (ER) membrane. In this study, we fused green fluorescent protein to the N or C terminus of human VKOR, expressed these fusions in HEK293 cells, and examined their topologies by fluorescence protease protection assays. Our results show that the N terminus of VKOR resides in the ER lumen, whereas its C terminus is in the cytoplasm. Selective modification of cysteines by polyethylene glycol maleimide confirms the cytoplasmic location of the conserved loop cysteines. Both results support a three-TMD model of VKOR. Interestingly, human VKOR can be changed to a four-TMD molecule by mutating the charged residues flanking the first TMD. Cell-based activity assays show that this four-TMD molecule is fully active. Furthermore, the conserved loop cysteines, which are essential for intramolecular electron transfer in the bacterial VKOR homologue, are not required for human VKOR whether they are located in the cytoplasm (three-TMD molecule) or the ER lumen (four-TMD molecule). Our results confirm that human VKOR is a three-TMD protein. Moreover, the conserved loop cysteines apparently play different roles in human VKOR and in its bacterial homologues.
- Published
- 2012
- Full Text
- View/download PDF
36. Mycobacterium tuberculosis vitamin K epoxide reductase homologue supports vitamin K-dependent carboxylation in mammalian cells.
- Author
-
Tie JK, Jin DY, and Stafford DW
- Subjects
- Cells, Cultured, Cysteine metabolism, HEK293 Cells, Humans, Vitamin K Epoxide Reductases, Mixed Function Oxygenases metabolism, Mycobacterium tuberculosis enzymology, Vitamin K metabolism
- Abstract
Aims: Vitamin K epoxide reductase complex, subunit 1 (VKORC1) is a critical participant in the production of active forms of reduced vitamin K and is required for modification of vitamin K-dependent proteins. Homologues of VKORC1 (VKORH) exist throughout evolution, but in bacteria they appear to function in oxidative protein folding as well as quinone reduction. In the current study we explore two questions: Do VKORHs function in the mammalian vitamin K cycle? Is the pair of loop cysteines-C43 and C51 in human VKORC1-conserved in all VKORC1s, essential for the activity of vitamin K epoxide reduction?, Results: We used our recently developed cell-based assay to compare the function of VKORHs to that of human VKORC1 in mammalian cells. We identified for the first time a VKORH (from Mycobacterium tuberculosis [Mt-VKORH]) that can function in the mammalian vitamin K cycle with vitamin K epoxide or vitamin K as substrate. Consistent with our previous in vitro results, the loop cysteines of human VKORC1 are not essential for its activity in vivo. Moreover, the corresponding loop cysteines of Mt-VKORH (C57 and C65), which are essential for its activity in disulfide bond formation during protein folding in Escherichia coli, are not required in the mammalian vitamin K cycle., Innovation and Conclusions: Our results indicate that VKORC1 in eukaryotes and Mt-VKORH in bacteria, that is, in their respective native environments, employ apparently different mechanisms for electron transfer. However, when Mt-VKORH is in the mammalian cell system, it employs a mechanism similar to that of VKORC1.
- Published
- 2012
- Full Text
- View/download PDF
37. Conformational transitions in the membrane scaffold protein of phospholipid bilayer nanodiscs.
- Author
-
Morgan CR, Hebling CM, Rand KD, Stafford DW, Jorgenson JW, and Engen JR
- Subjects
- 1,2-Dipalmitoylphosphatidylcholine chemistry, Apolipoprotein A-I metabolism, Deuterium metabolism, Deuterium Exchange Measurement, Humans, Hydrogen metabolism, Kinetics, Lipid Bilayers metabolism, Mass Spectrometry, Membrane Proteins metabolism, Membranes, Artificial, Models, Molecular, Molecular Conformation, Phospholipids metabolism, Protein Structure, Secondary, Protein Structure, Tertiary, Protein Unfolding, Recombinant Proteins metabolism, Solutions, Apolipoprotein A-I chemistry, Lipid Bilayers chemistry, Membrane Proteins chemistry, Nanostructures chemistry, Phospholipids chemistry, Proteomics methods, Recombinant Proteins chemistry
- Abstract
Phospholipid bilayer nanodiscs are model membrane systems that provide an environment where membrane proteins are highly stable and monodisperse without the use of detergents or liposomes. Nanodiscs consist of a discoidal phospholipid bilayer encircled by two copies of an amphipathic alpha helical membrane scaffold protein, which is modeled from apolipoprotein A-1. Hydrogen exchange mass spectrometry was used to probe the structure and dynamics of the scaffold protein in the presence and absence of lipid. On nanodisc self-assembly, the entire scaffold protein gained significant protection from exchange, consistent with a large, protein-wide, structural rearrangement. This protection was short-lived and the scaffold protein was highly deuterated within 2 h. Several regions of the scaffold protein, in both the lipid-free and lipid-associated states, displayed EX1 unfolding kinetics. The rapid deuteration of the scaffold protein and the presence of correlated unfolding events both indicate that nanodiscs are dynamic rather than rigid bodies in solution. This work provides a catalog of the expected scaffold protein peptic peptides in a nanodisc-hydrogen exchange mass spectrometry experiment and their deuterium uptake signatures, data that can be used as a benchmark to verify correct assembly and nanodisc structure. Such reference data will be useful control data for all hydrogen exchange mass spectrometry experiments involving nanodiscs in which transmembrane or lipid-associated proteins are the primary molecule(s) of interest.
- Published
- 2011
- Full Text
- View/download PDF
38. A hetero-dimer model for concerted action of vitamin K carboxylase and vitamin K reductase in vitamin K cycle.
- Author
-
Wu S, Liu S, Davis CH, Stafford DW, Kulman JD, and Pedersen LG
- Subjects
- Carbon-Carbon Ligases chemistry, Carbon-Carbon Ligases genetics, Catalytic Domain, Mutation genetics, NAD(P)H Dehydrogenase (Quinone) chemistry, NAD(P)H Dehydrogenase (Quinone) genetics, Protein Structure, Secondary, Quantum Theory, Warfarin pharmacology, Carbon-Carbon Ligases metabolism, Models, Biological, NAD(P)H Dehydrogenase (Quinone) metabolism, Protein Multimerization drug effects, Vitamin K metabolism
- Abstract
Vitamin K carboxylase (VKC) is believed to convert vitamin K, in the vitamin K cycle, to an alkoxide-epoxide form which then reacts with CO(2) and glutamate to generate γ-carboxyglutamic acid (Gla). Subsequently, vitamin K epoxide reductase (VKOR) is thought to convert the alkoxide-epoxide to a hydroquinone form. By recycling vitamin K, the two integral-membrane proteins, VKC and VKOR, maintain vitamin K levels and sustain the blood coagulation cascade. Unfortunately, NMR or X-ray crystal structures of the two proteins have not been characterized. Thus, our understanding of the vitamin K cycle is only partial at the molecular level. In this study, based on prior biochemical experiments on VKC and VKOR, we propose a hetero-dimeric form of VKC and VKOR that may explain the efficient oxidation and reduction of vitamin K during the vitamin K cycle., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
39. Functional study of the vitamin K cycle in mammalian cells.
- Author
-
Tie JK, Jin DY, Straight DL, and Stafford DW
- Subjects
- Anticoagulants pharmacology, Cell Line, Clinical Laboratory Techniques, HEK293 Cells, Humans, Mixed Function Oxygenases metabolism, Signal Transduction drug effects, Vitamin K 1 analogs & derivatives, Vitamin K 1 metabolism, Vitamin K Epoxide Reductases, Warfarin pharmacology, Signal Transduction physiology, Vitamin K metabolism
- Abstract
We describe a cell-based assay for studying vitamin K-cycle enzymes. A reporter protein consisting of the gla domain of factor IX (amino acids 1-46) and residues 47-420 of protein C was stably expressed in HEK293 and AV12 cells. Both cell lines secrete carboxylated reporter when fed vitamin K or vitamin K epoxide (KO). However, neither cell line carboxylated the reporter when fed KO in the presence of warfarin. In the presence of warfarin, vitamin K rescued carboxylation in HEK293 cells but not in AV12 cells. Dicoumarol, an NAD(P)H-dependent quinone oxidoreductase 1 (NQO1) inhibitor, behaved similarly to warfarin in both cell lines. Warfarin-resistant vitamin K epoxide reductase (VKOR-Y139F) supported carboxylation in HEK293 cells when fed KO in the presence of warfarin, but it did not in AV12 cells. These results suggest the following: (1) our cell system is a good model for studying the vitamin K cycle, (2) the warfarin-resistant enzyme reducing vitamin K to hydroquinone (KH₂) is probably not NQO1, (3) there appears to be a warfarin-sensitive enzyme other than VKOR that reduces vitamin K to KH₂, and (4) the primary function of VKOR is the reduction of KO to vitamin K.
- Published
- 2011
- Full Text
- View/download PDF
40. Quantum Chemical Study of the Mechanism of Action of Vitamin K Carboxylase in Solvent.
- Author
-
Wu S, Liu S, Davis CH, Stafford DW, and Pedersen LG
- Abstract
We investigate the post-translational generation of Gla (γ-carboxy glutamic acid) from Glu (glutamic acid) by vitamin K carboxylase (VKC) in solvent. VKC is thought to convert vitamin K, in the vitamin K cycle, to an alkoxide-epoxide form, which then reacts with CO(2) to generate an essential ingredient in blood coagulation, γ-carboxyglutamic acid (Gla). The generation of Gla from Glu is found to be exergenic (-15 kcal/mol) in aqueous solution with the SM6 method. We also produced the free energy profile for this model biochemical process with other solvent methods (polarizable continuum model, dielectric polarizable continuum model) and different dielectric constants. The biological implications are discussed.
- Published
- 2010
- Full Text
- View/download PDF
41. Effect of vitamin K-dependent protein precursor propeptide, vitamin K hydroquinone, and glutamate substrate binding on the structure and function of {gamma}-glutamyl carboxylase.
- Author
-
Higgins-Gruber SL, Mutucumarana VP, Lin PJ, Jorgenson JW, Stafford DW, and Straight DL
- Subjects
- Animals, Carbon-Carbon Ligases metabolism, Humans, Mice, Oligopeptides metabolism, Protein Precursors metabolism, Structure-Activity Relationship, Substrate Specificity, Tetraodontiformes, Vitamin K 2 metabolism, Carbon-Carbon Ligases chemistry, Oligopeptides chemistry, Protein Precursors chemistry, Vitamin K 2 chemistry
- Abstract
The γ-glutamyl carboxylase utilizes four substrates to catalyze carboxylation of certain glutamic acid residues in vitamin K-dependent proteins. How the enzyme brings the substrates together to promote catalysis is an important question in understanding the structure and function of this enzyme. The propeptide is the primary binding site of the vitamin K-dependent proteins to carboxylase. It is also an effector of carboxylase activity. We tested the hypothesis that binding of substrates causes changes to the carboxylase and in turn to the substrate-enzyme interactions. In addition we investigated how the sequences of the propeptides affected the substrate-enzyme interaction. To study these questions we employed fluorescently labeled propeptides to measure affinity for the carboxylase. We also measured the ability of several propeptides to increase carboxylase catalytic activity. Finally we determined the effect of substrates: vitamin K hydroquinone, the pentapeptide FLEEL, and NaHCO(3), on the stability of the propeptide-carboxylase complexes. We found a wide variation in the propeptide affinities for carboxylase. In contrast, the propeptides tested had similar effects on carboxylase catalytic activity. FLEEL and vitamin K hydroquinone both stabilized the propeptide-carboxylase complex. The two together had a greater effect than either alone. We conclude that the effect of propeptide and substrates on carboxylase controls the order of substrate binding in such a way as to ensure efficient, specific carboxylation.
- Published
- 2010
- Full Text
- View/download PDF
42. Conformational analysis of membrane proteins in phospholipid bilayer nanodiscs by hydrogen exchange mass spectrometry.
- Author
-
Hebling CM, Morgan CR, Stafford DW, Jorgenson JW, Rand KD, and Engen JR
- Subjects
- Deuterium Exchange Measurement, Deuterium Oxide chemistry, Hydrogen-Ion Concentration, Carbon-Carbon Ligases chemistry, Lipid Bilayers chemistry, Mass Spectrometry methods, Membrane Proteins chemistry, Nanostructures chemistry, Phospholipids chemistry
- Abstract
The study of membrane protein structure and enzymology has traditionally been hampered by the inherent insolubility of membrane proteins in aqueous environments and experimental challenges in emulating an in vivo lipid environment. Phospholipid bilayer nanodiscs have recently been shown to be of great use for the study of membrane proteins since they offer a controllable, stable, and monodisperse model membrane with a nativelike lipid bilayer. Here we report the integration of nanodiscs with hydrogen exchange (HX) mass spectrometry (MS) experiments, thereby allowing for analysis of the native conformation of membrane proteins. gamma-Glutamyl carboxylase (GGCX), an approximately 94 kDa transmembrane protein, was inserted into nanodiscs and labeled with deuterium oxide under native conditions. Analytical parameters including sample-handling and chromatographic separation were optimized to measure the incorporation of deuterium into GGCX. Coupling nanodisc technology with HX MS offers an effective approach for investigating the conformation and dynamics of membrane proteins in their native environment and is therefore capable of providing much needed insight into the function of membrane proteins.
- Published
- 2010
- Full Text
- View/download PDF
43. Abnormal hemostasis in a knock-in mouse carrying a variant of factor IX with impaired binding to collagen type IV.
- Author
-
Gui T, Reheman A, Ni H, Gross PL, Yin F, Monroe D, Monahan PE, and Stafford DW
- Subjects
- Animals, Binding Sites genetics, Factor IX analysis, Gene Knock-In Techniques, Hemophilia B, Hemorrhage, Mice, Protein Binding genetics, Thrombosis, Collagen Type IV metabolism, Factor IX genetics, Genetic Variation, Hemostasis genetics
- Abstract
Background: Factor IX binds to collagen type IV, but this binding has no known consequence., Objectives: To determine the effect of reduced binding of FIX to collagen IV., Methods: We constructed and characterized 'knock-in' mice containing the mutation lysine 5 to alanine (K5A) in the Gla domain of their FIX. The K5A mutation dramatically reduced the affinity of FIX for collagen type IV, but had no measurable effect on platelet binding, phospholipid binding, or in vitro clotting activity. However, K5AFIX mice had a mild bleeding tendency, despite their in vitro clotting activity being normal. Hemostatic protection from delayed rebleeding was intermediate between wild-type and hemophilia B mice (which had no detectable clotting activity); moreover, survival of K5A FIX mice after nascent clot removal was dramatically improved as compared with hemophilia B mice. Importantly, there was no detectable difference between K5AFIX and wild-type mice in either a laser-induced thrombosis model or the chromogenic FIX activity assay. In contrast, after ferric chloride injury, which exposes collagen IV as well as other basement membrane proteins, intravital microscopy revealed that vessel occlusion was significantly slower in K5AFIX mice than in wild-type mice., Conclusions: Our results indicate that the FIX molecule with decreased affinity for collagen IV has altered hemostatic properties in vivo and that the binding of FIX to collagen IV probably plays a significant functional role in hemostasis.
- Published
- 2009
- Full Text
- View/download PDF
44. Transmembrane domain interactions and residue proline 378 are essential for proper structure, especially disulfide bond formation, in the human vitamin K-dependent gamma-glutamyl carboxylase.
- Author
-
Tie JK, Zheng MY, Hsiao KL, Perera L, Stafford DW, and Straight DL
- Subjects
- Amino Acid Sequence, Amino Acid Substitution genetics, Carbon-Carbon Ligases genetics, Carbon-Carbon Ligases metabolism, Humans, Models, Molecular, Molecular Sequence Data, Peptide Fragments genetics, Peptide Fragments metabolism, Proline genetics, Protein Structure, Tertiary, Recombinant Proteins chemistry, Sequence Homology, Amino Acid, Structure-Activity Relationship, Carbon-Carbon Ligases chemistry, Cell Membrane enzymology, Disulfides chemistry, Peptide Fragments chemistry, Proline chemistry, Vitamin K chemistry
- Abstract
We used recombinant techniques to create a two-chain form (residues 1-345 and residues 346-758) of the vitamin K-dependent gamma-glutamyl carboxylase, a glycoprotein located in the endoplasmic reticulum containing five transmembrane domains. The two-chain carboxylase had carboxylase and epoxidase activities similar to those of one-chain carboxylase. In addition, it had normal affinity for the propeptide of factor IX. We employed this molecule to investigate formation of the one disulfide bond in carboxylase, the transmembrane structure of carboxylase, and the potential interactions among the carboxylase's transmembrane domains. Our results indicate that the two peptides of the two-chain carboxylase are joined by a disulfide bond. Proline 378 is important for the structure necessary for disulfide formation. Results with the P378L carboxylase indicate that noncovalent bonds maintain the two-chain structure even when the disulfide bond is disrupted. As we had previously proposed, the fifth transmembrane domain of carboxylase is the last and only transmembrane domain in the C-terminal peptide of the two-chain carboxylase. We show that the noncovalent association between the two chains of carboxylase involves an interaction between the fifth transmembrane domain and the second transmembrane domain. Results of a homology model of transmembrane domains 2 and 5 suggest that not only do these two domains associate but that transmembrane domain 2 may interact with another transmembrane domain. This latter interaction may be mediated at least in part by a motif of glycine residues in the second transmembrane domain.
- Published
- 2008
- Full Text
- View/download PDF
45. A quantum chemical study of the mechanism of action of Vitamin K epoxide reductase (VKOR) II. Transition states.
- Author
-
Davis CH, Deerfield D 2nd, Wymore T, Stafford DW, and Pedersen LG
- Subjects
- Epoxy Compounds chemistry, Mixed Function Oxygenases metabolism, Models, Chemical, Models, Molecular, Molecular Structure, Protons, Thermodynamics, Vitamin K chemistry, Vitamin K metabolism, Vitamin K Epoxide Reductases, Water chemistry, Mixed Function Oxygenases chemistry, Quantum Theory
- Abstract
A reaction path including transition states is generated for the Silverman mechanism [R.B. Silverman, Chemical model studies for the mechanism of Vitamin K epoxide reductase, J. Am. Chem. Soc. 103 (1981) 5939-5941] of action for Vitamin K epoxide reductase (VKOR) using quantum mechanical methods (B3LYP/6-311G**). VKOR, an essential enzyme in mammalian systems, acts to convert Vitamin K epoxide, formed by Vitamin K carboxylase, to its (initial) quinone form for cellular reuse. This study elaborates on a prior work that focused on the thermodynamics of VKOR [D.W. Deerfield II, C.H. Davis, T. Wymore, D.W. Stafford, L.G. Pedersen, Int. J. Quant. Chem. 106 (2006) 2944-2952]. The geometries of proposed model intermediates and transition states in the mechanism are energy optimized. We find that once a key disulfide bond is broken, the reaction proceeds largely downhill. An important step in the conversion of the epoxide back to the quinone form involves initial protonation of the epoxide oxygen. We find that the source of this proton is likely a free mercapto group rather than a water molecule. The results are consistent with the current view that the widely used drug Warfarin likely acts by blocking binding of Vitamin K at the VKOR active site and thereby effectively blocking the initiating step. These results will be useful for designing more complete QM/MM studies of the enzymatic pathway once three-dimensional structural data is determined and available for VKOR.
- Published
- 2007
- Full Text
- View/download PDF
46. A quantum chemical study of the mechanism of action of Vitamin K carboxylase (VKC) III. Intermediates and transition states.
- Author
-
Davis CH, Deerfield D 2nd, Wymore T, Stafford DW, and Pedersen LG
- Subjects
- Hydroquinones chemistry, Hydroquinones metabolism, Models, Chemical, Models, Molecular, Molecular Structure, Vitamin K chemistry, Vitamin K metabolism, Carbon-Carbon Ligases chemistry, Carbon-Carbon Ligases metabolism, Quantum Theory
- Abstract
A reaction path including transition states is generated for the Dowd mechanism [P. Dowd, R. Hershlne, S.W. Ham, S. Naganathan. Vitamin K and energy transduction: a base strength amplification mechanism. Science 269 (2005) 1684-1691] of action for Vitamin K carboxylase (VKC) using quantum chemical methods (B3LYP/6-311G**). VKC, an essential enzyme in mammalian systems, catalyzes the conversion of hydroquinone form of Vitamin K to the epoxide form in the presence of oxygen. An intermediate species of the oxidation of Vitamin K, an alkoxide, acts apparently to abstract the gamma hydrogen from specifically located glutamate residues. We are able to follow the Dowd proposed path to generate this alkoxide species. The geometries of the proposed model intermediates and transition states in the mechanism are energy optimized. We find that the most energetic step in the mechanism is the uni-deprotonation of the hydroquinone - once this occurs, there is only a small barrier of 3.5kcal/mol for the interaction of oxygen with the carbon to be attacked - and then the reaction proceeds downhill in free energy to form the critical alkoxide species. The results are consistent with the idea that the enzyme probably acts to facilitate the formation of the epoxide by reducing the energy required to deprotonate the hydroquinone form.
- Published
- 2007
- Full Text
- View/download PDF
47. Quantum chemical study of the mechanism of action of vitamin K carboxylase (VKC). IV. Intermediates and transition states.
- Author
-
Davis CH, Ii DD, Stafford DW, and Pedersen LG
- Subjects
- 1-Carboxyglutamic Acid biosynthesis, 1-Carboxyglutamic Acid chemical synthesis, Carbon Dioxide chemistry, Carbon Dioxide metabolism, Epoxy Compounds chemistry, Hydroquinones chemistry, Models, Biological, Molecular Structure, Vitamin K metabolism, Carbon-Carbon Ligases chemistry, Carbon-Carbon Ligases metabolism, Quantum Theory, Vitamin K chemistry
- Abstract
We studied proposed steps for the enzymatic formation of gamma-carboxyglutamic acid by density functional theory (DFT) quantum chemistry. Our results for one potentially feasible mechanism show that a vitamin K alkoxide intermediate can abstract a proton from glutamic acid at the gamma-carbon to form a carbanion and vitamin K epoxide. The hydrated carbanion can then react with CO2 to form gamma-carboxyglutamic acid. Computations at the B3LYP/6-311G** level were used to determine the intermediates and transition states for the overall process. The activation free energy for the gas-phase path is 22 kcal/mol, with the rate-limiting step for the reaction being the attack of the carbanion on CO2. Additional solvation studies, however, indicate that the formation of the carbanion step can be competitive with the CO2 attack step in high-dielectric systems. We relate these computations to the entire vitamin K cycle in the blood coagulation cascade, which is essential for viability of vertebrates.
- Published
- 2007
- Full Text
- View/download PDF
48. The conversion of vitamin K epoxide to vitamin K quinone and vitamin K quinone to vitamin K hydroquinone uses the same active site cysteines.
- Author
-
Jin DY, Tie JK, and Stafford DW
- Subjects
- Amino Acid Substitution, Catalytic Domain genetics, Cysteine chemistry, Disulfides chemistry, Enzyme Inhibitors pharmacology, Kinetics, Mixed Function Oxygenases antagonists & inhibitors, Mixed Function Oxygenases genetics, Mutagenesis, Site-Directed, Recombinant Proteins antagonists & inhibitors, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, Vitamin K 1 metabolism, Vitamin K Epoxide Reductases, Warfarin pharmacology, Mixed Function Oxygenases chemistry, Mixed Function Oxygenases metabolism, Vitamin K 1 analogs & derivatives, Vitamin K 2 metabolism
- Abstract
Vitamin K epoxide (or oxido) reductase (VKOR) is the target of warfarin and provides vitamin K hydroquinone for the carboxylation of select glutamic acid residues of the vitamin K-dependent proteins which are important for coagulation, signaling, and bone metabolism. It has been known for at least 20 years that cysteines are required for VKOR function. To investigate their importance, we mutated each of the seven cysteines in VKOR. In addition, we made VKOR with both C43 and C51 mutated to alanine (C43A/C51A), as well as a VKOR with residues C43-C51 deleted. Each mutated enzyme was purified and characterized. We report here that C132 and C135 of the CXXC motif are essential for both the conversion of vitamin K epoxide to vitamin K and the conversion of vitamin K to vitamin K hydroquinone. Surprisingly, conserved cysteines, 43 and 51, appear not to be important for either reaction. For the in vitro reaction driven by dithiothreitol, the 43-51 deletion mutation retained 85% and C43A/C51A 112% of the wild-type activity. The facile purification of the nine different mutations reported here illustrates the ease and reproducibility of VKOR purification by the method reported in our recent publication [Chu, P.-H., Huang, T.-Y., Williams, J., and Stafford, D. W. (2006) Proc. Natl. Acad. Sci. U S A. 103, 19308-19313].
- Published
- 2007
- Full Text
- View/download PDF
49. Vitamin K supplementation during oral anticoagulation: concerns.
- Author
-
Stafford DW, Roberts HR, and Vermeer C
- Subjects
- Aged, Aged, 80 and over, Anticoagulants pharmacology, Blood Coagulation Disorders blood, Drug Stability, Female, Humans, Male, Middle Aged, Vitamin K administration & dosage, Vitamin K blood, Warfarin pharmacology, Anticoagulants therapeutic use, Blood Coagulation Disorders drug therapy, Dietary Supplements, Vitamin K pharmacology, Warfarin therapeutic use
- Published
- 2007
- Full Text
- View/download PDF
50. Transgene expression levels and kinetics determine risk of humoral immune response modeled in factor IX knockout and missense mutant mice.
- Author
-
Zhang TP, Jin DY, Wardrop RM 3rd, Gui T, Maile R, Frelinger JA, Stafford DW, and Monahan PE
- Subjects
- Animals, Cross Reactions, Factor IX immunology, Gene Expression, Genetic Therapy methods, Hemophilia A blood, Hemophilia A immunology, Hemophilia A therapy, Humans, Kinetics, Mice, Mice, Knockout, Mice, Mutant Strains, Models, Biological, Mutation, Missense, Risk, Transduction, Genetic methods, Transgenes, Autoantibodies immunology, Factor IX genetics, Genetic Therapy adverse effects, Genetic Vectors genetics
- Abstract
Immune responses leading to antibody-mediated elimination of the transgenic protein are a concern in gene replacement for congenital protein deficiencies, for which hemophilia is an important model. Although most hemophilia B patients have circulating non-functional but immunologically crossreactive factor IX (FIX) protein (CRM+ phenotype), inciting factors for FIX neutralizing antibody (inhibitor) development have been studied in crossreactive material-negative (CRM-) animal models. For this study, determinants of FIX inhibitor development were compared in hemophilia B mice, in which circulating FIX protein is absent (CRM- factor IX knockout (FIXKO) model) or present (CRM+ missense R333Q-hFIX model) modeling multiple potential therapies. The investigations compare for the first time different serotypes of adeno-associated virus (AAV) vectors (AAV2 and AAV1), each at multiple doses, in the setting of two different FIX mutations. The comparisons demonstrate in the FIXKO background (CRM- phenotype) that neither vector serotype nor vector particle number independently determine the inhibitor trigger, which is influenced primarily by the level and kinetics of transgene expression. In the CRM+ missense background, inhibitor development was never stimulated by AAV gene therapy or protein therapy, despite the persistence of lymphocytes capable of responding to FIX with non-inhibitory antibodies. This genotype/phenotype is strongly protective against antibody formation in response to FIX therapy.
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.