Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nutrients ; 7(8): 6250-80, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26264021

ABSTRACT

Among all cellular life on earth, with the exception of yeasts, fungi, and some prokaryotes, VKOR family homologs are ubiquitously encoded in nuclear genomes, suggesting ancient and important biological roles for these enzymes. Despite single gene and whole genome duplications on the largest evolutionary timescales, and the fact that most gene duplications eventually result in loss of one copy, it is surprising that all jawed vertebrates (gnathostomes) have retained two paralogous VKOR genes. Both VKOR paralogs function as entry points for nutritionally acquired and recycled K vitamers in the vitamin K cycle. Here we present phylogenetic evidence that the human paralogs likely arose earlier than gnathostomes, possibly in the ancestor of crown chordates. We ask why gnathostomes have maintained these paralogs throughout evolution and present a current summary of what we know. In particular, we look to published studies about tissue- and developmental stage-specific expression, enzymatic function, phylogeny, biological roles and associated pathways that together suggest subfunctionalization as a major influence in evolutionary fixation of both paralogs. Additionally, we investigate on what evolutionary timescale the paralogs arose and under what circumstances in order to gain insight into the biological raison d'être for both VKOR paralogs in gnathostomes.


Subject(s)
Biological Evolution , Phylogeny , Vertebrates/genetics , Vitamin K Epoxide Reductases/genetics , Vitamin K/metabolism , Animals , Humans , Sequence Homology, Amino Acid , Vitamin K Epoxide Reductases/metabolism
2.
Nutrients ; 7(8): 6224-49, 2015 Jul 29.
Article in English | MEDLINE | ID: mdl-26230708

ABSTRACT

In humans and other vertebrate animals, vitamin K 2,3-epoxide reductase (VKOR) family enzymes are the gatekeepers between nutritionally acquired K vitamins and the vitamin K cycle responsible for posttranslational modifications that confer biological activity upon vitamin K-dependent proteins with crucial roles in hemostasis, bone development and homeostasis, hormonal carbohydrate regulation and fertility. We report a phylogenetic analysis of the VKOR family that identifies five major clades. Combined phylogenetic and site-specific conservation analyses point to clade-specific similarities and differences in structure and function. We discovered a single-site determinant uniquely identifying VKOR homologs belonging to human pathogenic, obligate intracellular prokaryotes and protists. Building on previous work by Sevier et al. (Protein Science 14:1630), we analyzed structural data from both VKOR and prokaryotic disulfide bond formation protein B (DsbB) families and hypothesize an ancient evolutionary relationship between the two families where one family arose from the other through a gene duplication/deletion event. This has resulted in circular permutation of primary sequence threading through the four-helical bundle protein folds of both families. This is the first report of circular permutation relating distant a-helical membrane protein sequences and folds. In conclusion, we suggest a chronology for the evolution of the five extant VKOR clades.


Subject(s)
Biological Evolution , Disulfides , Phylogeny , Receptors, Fc/genetics , Vitamin K Epoxide Reductases/genetics , Vitamin K/metabolism , Amino Acid Sequence , Animals , Archaea , Bacteria , Humans , Molecular Sequence Data , Protein Structure, Secondary , Sequence Homology, Amino Acid , Vitamin K Epoxide Reductases/chemistry
3.
Anal Biochem ; 474: 89-94, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25524619

ABSTRACT

Use of the reductant dithiothreitol (DTT) as a substrate for measuring vitamin K 2,3-epoxide reductase (VKOR) activity in vitro has been reported to be problematic because it enables side reactions involving the vitamin K1 2,3-epoxide (K1>O) substrate. Here we characterize specific problems when using DTT and show that tris(3-hydroxypropyl)phosphine (THPP) is a reliable alternative to DTT for in vitro assessment of VKOR enzymatic activity. In addition, the pH buffering compound imidazole was found to be problematic in enhancing DTT-dependent non-enzymatic side reactions. Using THPP and phosphate-based pH buffering, we measured apparent Michaelis-Menten constants of 1.20 µM for K1>O and 260 µM for the active neutral form of THPP. The Km value for K1>O is in agreement with the value that we previously obtained using DTT (1.24 µM). Using THPP, we successfully eliminated non-enzymatic production of 3-hydroxyvitamin K1 and its previously reported base-catalyzed conversion to K1, both of which were shown to occur when DTT and imidazole are used as the reductant and pH buffer, respectively, in the in vitro VKOR assay. Accordingly, substitution of THPP for DTT in the in vitro VKOR assay will ensure more accurate enzymatic measurements and assessment of warfarin and other 4-hydroxycoumarin inhibition constants.


Subject(s)
Dithiothreitol/metabolism , Phosphines/metabolism , Vitamin K Epoxide Reductases/metabolism , Biocatalysis , Buffers , Cholic Acids/metabolism , Enzyme Assays , Humans , Hydrogen-Ion Concentration , Kinetics , Oxidation-Reduction , Pichia/metabolism , Reducing Agents/metabolism , Solutions , Substrate Specificity
4.
Hum Mutat ; 34(11): 1490-500, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23913518

ABSTRACT

Coagulation factor XIII (FXIII) proenzyme circulates in plasma as a heterotetramer composed of two each of A and B subunits. Upon activation, the B subunits dissociate from the A subunit dimer, which gains transglutaminase activity to cross-link preformed fibrin clots increasing mechanical strength and resistance to degradation. The B subunits are thought to possess a carrier/protective function before FXIII activation. Mutations in either A or B subunits are associated with pathological patient phenotypes characterized by mild to severe bleeding. In vitro expression of FXIII B subunit (FXIIIB) missense variants in HEK293T cells revealed impaired secretion for all seven variants studied. To investigate the likely molecular environments of the missense residues, we created molecular models of individual FXIIIB Sushi domains using phylogenetically similar complement factor H Sushi domain structural templates. Assessment of the local molecular environments for the models suggested surface or buried positions for each mutant residue and possible pathological mechanisms. The in vitro expression system and in silico analytical methods and models we developed can be used to further investigate the molecular basis of FXIIIB mutation pathologies.


Subject(s)
Factor XIII/genetics , Mutation, Missense , Phenotype , Cell Line , Factor XIII/chemistry , Factor XIII/metabolism , Gene Expression , Heterozygote , Humans , Models, Molecular , Protein Conformation , Protein Interaction Domains and Motifs
5.
Biochim Biophys Acta ; 1830(8): 4202-10, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23618698

ABSTRACT

BACKGROUND: Warfarin directly inhibits vitamin K 2,3-epoxide reductase (VKOR) enzymes. Since the early 1970s, warfarin inhibition of vitamin K 2,3-epoxide reductase complex subunit 1 (VKORC1), an essential enzyme for proper function of blood coagulation in higher vertebrates, has been studied using an in vitro dithiothreitol (DTT) driven enzymatic assay. However, various studies based on this assay have reported warfarin dose-response data, usually summarized as half-maximal inhibitory concentration (IC50), that vary over orders of magnitude and reflect the broad range of conditions used to obtain VKOR assay data. METHODS: We standardized the implementation of the DTT-driven VKOR activity assay to measure enzymatic Michaelis constants (Km) and warfarin IC50 for human VKORC1. A data transformation is defined, based on the previously confirmed bi bi ping-pong mechanism for VKORC1, that relates assay condition-dependent IC50 to condition-independent Ki. RESULTS: Determination of the warfarin Ki specifically depends on measuring both substrate concentrations, both Michaelis constants for the VKORC1 enzyme, and pH in the assay. CONCLUSION: The Ki is not equal to the IC50 value directly measured using the DTT-driven VKOR assay. GENERAL SIGNIFICANCE: In contrast to warfarin IC50 values determined in previous studies, warfarin inhibition expressed as Ki can now be compared between studies, even when the specific DTT-driven VKOR assay conditions differ. This implies that warfarin inhibition reported for wild-type and variant VKORC1 enzymes from previous reports should be reassessed and new determinations of Ki are required to accurately report and compare in vitro warfarin inhibition results.


Subject(s)
Dithiothreitol/pharmacology , Mixed Function Oxygenases/antagonists & inhibitors , Warfarin/pharmacology , Humans , Kinetics , Vitamin K Epoxide Reductases
6.
Methods Enzymol ; 520: 1-29, 2013.
Article in English | MEDLINE | ID: mdl-23332693

ABSTRACT

Recent success in obtaining high-resolution structural data for the first several G protein-coupled receptors (GPCRs) has highlighted the feasibility of structural membrane proteomics approaches for obtaining molecular models of additional GPCRs from among the nearly 800 encoded by the human genome. Yet, production of functional receptors, in general, has proven to be difficult, typically requiring considerable time and cost investments. Here we describe screening, optimization, and scale-up methods we successfully used to produce milligram amounts of functional GPCRs in Pichia pastoris. When we surveyed a large number of receptors recombinantly produced in Pichia, 85% exhibited specific ligand binding, strongly suggesting that this expression system is excellent for producing functional GPCRs. Of the latter group, 20 were optimized according to our protocol. Of these, we produced 10 as milligram amounts of functional receptors using large-scale shaker culture. Cost and time expenditures were considerably lower using the Pichia system than for other successfully employed cell culture systems.


Subject(s)
Pichia/metabolism , Receptors, G-Protein-Coupled/metabolism , Pichia/genetics , Receptors, G-Protein-Coupled/genetics , Transformation, Genetic
7.
J Biol Chem ; 286(17): 15085-94, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21367861

ABSTRACT

Human vitamin K 2,3-epoxide reductase complex subunit 1-like 1 (VKORC1L1), expressed in HEK 293T cells and localized exclusively to membranes of the endoplasmic reticulum, was found to support both vitamin K 2,3-epoxide reductase (VKOR) and vitamin K reductase enzymatic activities. Michaelis-Menten kinetic parameters for dithiothreitol-driven VKOR activity were: K(m) (µM) = 4.15 (vitamin K(1) epoxide) and 11.24 (vitamin K(2) epoxide); V(max) (nmol·mg(-1)·hr(-1)) = 2.57 (vitamin K(1) epoxide) and 13.46 (vitamin K(2) epoxide). Oxidative stress induced by H(2)O(2) applied to cultured cells up-regulated VKORC1L1 expression and VKOR activity. Cell viability under conditions of no induced oxidative stress was increased by the presence of vitamins K(1) and K(2) but not ubinquinone-10 and was specifically dependent on VKORC1L1 expression. Intracellular reactive oxygen species levels in cells treated with 2,3-dimethoxy-1,4-naphthoquinone were mitigated in a VKORC1L1 expression-dependent manner. Intracellular oxidative damage to membrane intrinsic proteins was inversely dependent on VKORC1L1 expression and the presence of vitamin K(1). Taken together, our results suggest that VKORC1L1 is responsible for driving vitamin K-mediated intracellular antioxidation pathways critical to cell survival.


Subject(s)
Antioxidants/metabolism , Mixed Function Oxygenases/metabolism , Cell Line , Cell Survival , Endoplasmic Reticulum/metabolism , Humans , Hydrogen Peroxide , Intracellular Space/metabolism , Kinetics , Mixed Function Oxygenases/biosynthesis , Mixed Function Oxygenases/genetics , Oxidation-Reduction , Oxidative Stress , Protein Subunits , Vitamin K 1 , Vitamin K Epoxide Reductases
8.
Thromb Haemost ; 98(3): 570-8, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17849045

ABSTRACT

For decades coumarins have been the most commonly prescribed drugs for therapy and prophylaxis of thromboembolic conditions. Despite the limitation of their narrow therapeutic dosage window, the broad variation of intra- and inter-individual drug requirement, and the relatively high incidence of bleeding complications, prescriptions for coumarins are increasing due to the aging populations in industrialised countries. The identification of the molecular target of coumarins, VKORC1, has greatly improved the understanding of coumarin treatment and illuminated new perspectives for a safer and more individualized oral anticoagulation therapy. Mutations and SNPs within the translated and non-translated regions of the VKORC1 gene have been shown to cause coumarin resistance and sensitivity, respectively. Besides the known CYP2C9 variants that affect coumarin metabolism, the haplotype VKORC1*2 representing a frequent SNP within the VKORC1 promoter has been identified as a major determinant of coumarin sensitivity, reducing VKORC1 enzyme activity to 50% of wild type. Homozygous carriers of the VKORC1*2 allele are strongly predisposed to coumarin sensitivity. Using individualized dose adaptation, a significant reduction of bleeding complications can be expected, especially in the initial drug saturation phase. Furthermore, concomitant application of low dose vitamin K may significantly reduce intra-individual coumarin dose variation and, thus, may stabilize oral anticoagulation therapy. The use of new pharmacogenetics-based dosing schemes and the concomitant application of low-dose vitamin K with coumarins will decidedly influence the current practice of oral anticoagulation and greatly improve coumarin drug safety.


Subject(s)
Anticoagulants/administration & dosage , Aryl Hydrocarbon Hydroxylases/genetics , Coumarins/administration & dosage , Hemorrhage/chemically induced , Mixed Function Oxygenases/genetics , Mutation , Pharmacogenetics/trends , Polymorphism, Single Nucleotide , Administration, Oral , Algorithms , Anticoagulants/adverse effects , Anticoagulants/metabolism , Antifibrinolytic Agents/administration & dosage , Antifibrinolytic Agents/metabolism , Aryl Hydrocarbon Hydroxylases/metabolism , Coumarins/adverse effects , Coumarins/metabolism , Cytochrome P-450 CYP2C9 , Dose-Response Relationship, Drug , Drug Resistance , Drug Therapy, Combination , Gene Frequency , Genetic Predisposition to Disease , Genotype , Haplotypes , Hemorrhage/enzymology , Hemorrhage/metabolism , Hemorrhage/prevention & control , Humans , Mixed Function Oxygenases/chemistry , Mixed Function Oxygenases/metabolism , Phenotype , Protein Conformation , Risk Assessment , Risk Factors , Vitamin K/administration & dosage , Vitamin K/metabolism , Vitamin K Epoxide Reductases
9.
Antioxid Redox Signal ; 8(3-4): 347-53, 2006.
Article in English | MEDLINE | ID: mdl-16677080

ABSTRACT

Vitamin K epoxide, a by-product of the carboxylation of blood coagulation factors, is reduced to vitamin K by an enzymatic system possessing vitamin K epoxide reductase (VKOR) activity. This system is the target of coumarin-derived drugs widely used in thrombosis therapy and prophylaxis. Recently, the key protein of the VKOR system has been identified. The human VKORC1 gene maps to chromosome 16 and consists of 3 exons encoding a 163-amino acid integral ER membrane protein with three or four predicted transmembrane alpha- helices. Expression of human VKORC1 in Spodoptera frugiperda (Sf9) cells and in Pichia pastoris results in enhanced VKOR activity over low endogenous constitutive levels. Sequence based search methods reveal that human VKORC1 belongs to a large family of homologous genes found in vertebrates, insects, plants, protists, archea, and bacteria. All orthologs share five completely conserved amino acids, including two cysteines found in a tetrapeptide motif presumably required for redox function. The recent discovery of the VKORC1 gene has initiated renewed interest in understanding VKOR activity. Analysis of VKORC1 protein structure and function will be crucial in understanding the VKOR catalytic mechanism, how anticoagulant drugs modulate VKOR activity, and the role of VKORC1 in downstream physiological and pathological pathways.


Subject(s)
Mixed Function Oxygenases/physiology , Vitamin K/physiology , Amino Acid Sequence , Animals , Coumarins/chemistry , Cysteine/chemistry , Gene Expression Regulation , Gene Expression Regulation, Enzymologic , Humans , Mixed Function Oxygenases/metabolism , Molecular Sequence Data , Pichia/metabolism , Protein Conformation , Spodoptera/metabolism , Thrombosis/metabolism , Vitamin K/chemistry , Vitamin K Epoxide Reductases
10.
Thromb Haemost ; 94(4): 780-6, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16270630

ABSTRACT

Coumarin and homologous compounds are the most widely used anticoagulant drugs worldwide. They function as antagonists of vitamin K, an essential cofactor for the posttranslational gamma-glutamyl carboxylation of the so-called vitamin K-dependent proteins. As vitamin K hydroquinone is converted to vitamin K epoxide (VKO) in every carboxylation step, the epoxide has to be recycled to the reduced form by the vitamin K epoxide reductase complex (VKOR). Recently, a single coumarin-sensitive protein of the putative VKOR enzyme complex was identified in humans (vitamin K epoxide reductase complex subunit 1, VKORC1). Mutations in VKORC1 result in two different phenotypes: warfarin resistance (WR) and multiple coagulation factor deficiency type 2 (VKCFD2). Here,we report on the expression of site-directed VKORC1 mutants, addressing possible structural and functional roles of all seven cysteine residues (Cys16, Cys43, Cys51, Cys85, Cys96, Cys132, Cys135), the highly conserved residue Ser/Thr57, and Arg98, known to cause VKCFD2 in humans. Our results support the hypothesis that the C132-X-X-C135 motif in VKORC1 comprises part of the redox active site that catalyzes VKO reduction and also suggest a crucial role for the hydrophobic Thr-Tyr-Ala motif in coumarin binding. Furthermore, our results support the concept that different structural components of VKORC1 define the binding sites for vitamin K epoxide and coumarin.


Subject(s)
Anticoagulants/pharmacology , Coumarins/pharmacology , Drug Resistance/genetics , Mixed Function Oxygenases/genetics , Amino Acid Sequence , Anticoagulants/metabolism , Binding Sites/genetics , Cell Line , Coumarins/metabolism , Disulfides/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Kidney/cytology , Mixed Function Oxygenases/chemistry , Mixed Function Oxygenases/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Vitamin K 1/analogs & derivatives , Vitamin K 1/metabolism , Vitamin K Epoxide Reductases
11.
Carbohydr Res ; 339(2): 221-31, 2004 Jan 22.
Article in English | MEDLINE | ID: mdl-14698880

ABSTRACT

To facilitate the use of oligosaccharides as analytical tools in biological studies, we have designed, synthesized, and conjugated to maltosaccharides a novel series of homologous small fluorescent moieties that differ in formal charge. These moieties are amide derivatives of anthranilic acid: uncharged N-(2-aminobenzoyl)glycinamide (ABGlyAmide; 2), acidic N,N-dimethyl-N(')-(2-aminobenzoyl)ethylenediamine (ABGlyDIMED; 3), and basic N-(2-aminobenzoyl)glycine (ABGly; 1). Routes for synthesis and optimal reaction conditions for glycoconjugation by conventional reductive amination are presented, as is the compatibility of these adducts with common analytical and preparative chromatographic methods, including RP-HPLC and HPAEC-PAD. These novel anthranilic acid derivatives confer both fluorescence and defined charge to oligosaccharides, and so enhance the repertoire of chromatographic and analytical methods for which anthranilic acid can be used. Furthermore, because glucosaccharides have rigid solution structure, these small fluorescent adducts with different formal charge are ideal tools for molecular sizing studies of membrane pores.


Subject(s)
Oligosaccharides/chemistry , ortho-Aminobenzoates/chemistry , Acids/chemistry , Adsorption , Amines/chemistry , Carbohydrate Conformation , Hydrogen-Ion Concentration , Ions/chemistry , Molecular Structure , Oligosaccharides/isolation & purification , Oxidation-Reduction , Solutions/chemistry , Spectrum Analysis , Static Electricity , ortho-Aminobenzoates/chemical synthesis
12.
Cell Commun Adhes ; 10(4-6): 239-44, 2003.
Article in English | MEDLINE | ID: mdl-14681023

ABSTRACT

The mechanisms of molecular discrimination by connexin channels are of acute biological and medical importance. The availability of affinity or open-pore blocking reagents for reliable and specific study of the connexin permeability pathway, would make possible the rigorous cellular and physiological studies required to inform, in molecular terms, the underlying role of intercellular communication pathways in development and disease. Previous work utilized a series of glucosaccharides labeled with an uncharged fluorescent aminopyridine (PA-) group to establish steric constraints to permeability through connexin hemichannels. In that work, the smallest probe permeable through homomeric Cx26 and heteromeric Cx26-Cx32 channels was the PA-disaccharide, and the smallest probe permeable through homomeric Cx32 channels was the PA-trisaccharide. The larger impermeable probes did not block permeation of the smaller probes. Building on this work, a new set of glucosaccharide probes was developed in which the label was one of a homologous series of novel anthranilic acid derivatives (ABG) that carry negative or positive formal charge or remain neutral at physiological pH. When the PA-label of the smallest impermeant PA-derivatized oligosaccharides was replaced by ABG label, the resulting probes acted as reversible, high-affinity inhibitors of large molecule permeation through connexin pores in a size and connexin-specific manner.


Subject(s)
Cell Membrane Permeability/drug effects , Connexins/metabolism , ortho-Aminobenzoates/pharmacology , Animals , Connexin 26 , Fluorescent Dyes/chemistry , Glycoconjugates/pharmacology , Hepatocytes/metabolism , Humans , Mammary Glands, Human/metabolism , ortho-Aminobenzoates/analysis , Gap Junction beta-1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...