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










Publication year range
1.
Proc Natl Acad Sci U S A ; 105(11): 4115-20, 2008 Mar 18.
Article in English | MEDLINE | ID: mdl-18332423

ABSTRACT

B(12)-dependent methionine synthase (MetH) from Escherichia coli is a large modular protein that is alternately methylated by methyltetrahydrofolate to form methylcobalamin and demethylated by homocysteine to form cob(I)alamin. Major domain rearrangements are required to allow cobalamin to react with three different substrates: homocysteine, methyltetrahydrofolate, and S-adenosyl-l-methionine (AdoMet). These same rearrangements appear to preclude crystallization of the wild-type enzyme. Disulfide cross-linking was used to lock a C-terminal fragment of the enzyme into a unique conformation. Cysteine point mutations were introduced at Ile-690 and Gly-743. These cysteine residues span the cap and the cobalamin-binding module and form a cross-link that reduces the conformational space accessed by the enzyme, facilitating protein crystallization. Here, we describe an x-ray structure of the mutant fragment in the reactivation conformation; this conformation enables the transfer of a methyl group from AdoMet to the cobalamin cofactor. In the structure, the axial ligand to the cobalamin, His-759, dissociates from the cobalamin and forms intermodular contacts with residues in the AdoMet-binding module. This unanticipated intermodular interaction is expected to play a major role in controlling the distribution of conformers required for the catalytic and the reactivation cycles of the enzyme.


Subject(s)
5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/chemistry , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/metabolism , Disulfides/chemistry , Disulfides/metabolism , Histidine/metabolism , Vitamin B 12/chemistry , Vitamin B 12/metabolism , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/genetics , Binding Sites , Catalysis , Crystallography, X-Ray , Enzyme Activation , Escherichia coli/enzymology , Escherichia coli/genetics , Gene Expression , Histidine/genetics , Ligands , Models, Molecular , Photochemistry , Protein Structure, Quaternary , Protein Structure, Tertiary , Structural Homology, Protein , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/metabolism , Temperature , Titrimetry
2.
Proc Natl Acad Sci U S A ; 105(9): 3286-91, 2008 Mar 04.
Article in English | MEDLINE | ID: mdl-18296644

ABSTRACT

Enzymes possessing catalytic zinc centers perform a variety of fundamental processes in nature, including methyl transfer to thiols. Cobalamin-independent (MetE) and cobalamin-dependent (MetH) methionine synthases are two such enzyme families. Although they perform the same net reaction, transfer of a methyl group from methyltetrahydrofolate to homocysteine (Hcy) to form methionine, they display markedly different catalytic strategies, modular organization, and active site zinc centers. Here we report crystal structures of zinc-replete MetE and MetH, both in the presence and absence of Hcy. Structural investigation of the catalytic zinc sites of these two methyltransferases reveals an unexpected inversion of zinc geometry upon binding of Hcy and displacement of an endogenous ligand in both enzymes. In both cases a significant movement of the zinc relative to the protein scaffold accompanies inversion. These structures provide new information on the activation of thiols by zinc-containing enzymes and have led us to propose a paradigm for the mechanism of action of the catalytic zinc sites in these and related methyltransferases. Specifically, zinc is mobile in the active sites of MetE and MetH, and its dynamic nature helps facilitate the active site conformational changes necessary for thiol activation and methyl transfer.


Subject(s)
5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/chemistry , Homocysteine/metabolism , Metals , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/metabolism , Binding Sites , Catalysis , Crystallography, X-Ray , Elasticity , Methylation , Methyltransferases , Protein Conformation , Thermotoga maritima/enzymology , Vitamin B 12 , Zinc
3.
Biochemistry ; 45(15): 4808-18, 2006 Apr 18.
Article in English | MEDLINE | ID: mdl-16605249

ABSTRACT

In human methylenetetrahydrofolate reductase (MTHFR) the Ala222Val (677C-->T) polymorphism encodes a heat-labile gene product that is associated with elevated levels of homocysteine and possibly with risk for cardiovascular disease. Generation of the equivalent Ala to Val mutation in Escherichia coli MTHFR, which is 30% identical to the catalytic domain of the human enzyme, creates a protein with enhanced thermolability. In both human and E. coli MTHFR, the A --> V mutation increases the rate of dissociation of FAD, and in both enzymes, loss of FAD is linked to changes in quaternary structure [Yamada, K., Chen, Z., Rozen, R., and Matthews, R. G. (2001) Proc. Natl. Acad. Sci. U.S.A. 98, 14853-14858; Guenther, B. D., Sheppard, C. A., Tran, P., Rozen, R., Matthews, R. G., and Ludwig, M. L. (1999) Nat. Struct. Biol. 6, 359-365]. Folates have been shown to protect both human and bacterial enzymes from loss of FAD. Despite its effect on affinity for FAD, the A --> V mutation is located at the bottom of the (betaalpha)(8) barrel of the catalytic domain in a position that does not contact the bound FAD prosthetic group. Here we report the structures of the Ala177Val mutant of E. coli MTHFR and of its complex with the 5,10-dideazafolate analogue, LY309887, and suggest mechanisms by which the mutation may perturb FAD binding. Helix alpha5, which immediately precedes the loop bearing the mutation, carries several residues that interact with FAD, including Asn168, Arg171, and Lys172. In the structures of the mutant enzyme this helix is displaced, perturbing protein-FAD interactions. In the complex with LY309887, the pterin-like ring of the analogue stacks against the si face of the flavin and is secured by hydrogen bonds to residues Gln183 and Asp120 that adjoin this face. The direct interactions of bound folate with the cofactor provide one mechanism for linkage between binding of FAD and folate binding that could account in part for the protective action of folates. Conformation changes induced by folate binding may also suppress dissociation of FAD.


Subject(s)
Alanine/genetics , Enzyme Activation , Folic Acid/metabolism , Polymorphism, Genetic , Protein Structure, Secondary , Valine/genetics , Alanine/metabolism , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Enzyme Activation/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli/metabolism , Flavin-Adenine Dinucleotide/chemistry , Flavin-Adenine Dinucleotide/metabolism , Humans , Ligands , Methylenetetrahydrofolate Reductase (NADPH2) , Models, Molecular , Molecular Sequence Data , Mutation , Polymorphism, Genetic/physiology , Protein Binding , Protein Conformation , Protein Structure, Secondary/genetics , Structure-Activity Relationship , Valine/metabolism
4.
Biochemistry ; 44(34): 11447-57, 2005 Aug 30.
Article in English | MEDLINE | ID: mdl-16114881

ABSTRACT

Methylenetetrahydrofolate reductases (MTHFRs; EC 1.7.99.5) catalyze the NAD(P)H-dependent reduction of 5,10-methylenetetrahydrofolate (CH(2)-H(4)folate) to 5-methyltetrahydrofolate (CH(3)-H(4)folate) using flavin adenine dinucleotide (FAD) as a cofactor. The initial X-ray structure of Escherichia coli MTHFR revealed that this 33-kDa polypeptide is a (betaalpha)(8) barrel that aggregates to form an unusual tetramer with only 2-fold symmetry. Structures of reduced enzyme complexed with NADH and of oxidized Glu28Gln enzyme complexed with CH(3)-H(4)folate have now been determined at resolutions of 1.95 and 1.85 A, respectively. The NADH complex reveals a rare mode of dinucleotide binding; NADH adopts a hairpin conformation and is sandwiched between a conserved phenylalanine, Phe223, and the isoalloxazine ring of FAD. The nicotinamide of the bound pyridine nucleotide is stacked against the si face of the flavin ring with C4 adjoining the N5 of FAD, implying that this structure models a complex that is competent for hydride transfer. In the complex with CH(3)-H(4)folate, the pterin ring is also stacked against FAD in an orientation that is favorable for hydride transfer. Thus, the binding sites for the two substrates overlap, as expected for many enzymes that catalyze ping-pong reactions, and several invariant residues interact with both folate and pyridine nucleotide substrates. Comparisons of liganded and substrate-free structures reveal multiple conformations for the loops beta2-alpha2 (L2), beta3-alpha3 (L3), and beta4-alpha4 (L4) and suggest that motions of these loops facilitate the ping-pong reaction. In particular, the L4 loop adopts a "closed" conformation that allows Asp120 to hydrogen bond to the pterin ring in the folate complex but must move to an "open" conformation to allow NADH to bind.


Subject(s)
Escherichia coli/enzymology , Folic Acid/metabolism , Methylenetetrahydrofolate Reductase (NADPH2)/chemistry , Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , NAD/metabolism , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Hydrogen-Ion Concentration , Kinetics , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Structure, Secondary
5.
PLoS Biol ; 3(2): e31, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15630480

ABSTRACT

Cobalamin-independent methionine synthase (MetE) catalyzes the transfer of a methyl group from methyltetrahydrofolate to L-homocysteine (Hcy) without using an intermediate methyl carrier. Although MetE displays no detectable sequence homology with cobalamin-dependent methionine synthase (MetH), both enzymes require zinc for activation and binding of Hcy. Crystallographic analyses of MetE from T. maritima reveal an unusual dual-barrel structure in which the active site lies between the tops of the two (betaalpha)(8) barrels. The fold of the N-terminal barrel confirms that it has evolved from the C-terminal polypeptide by gene duplication; comparisons of the barrels provide an intriguing example of homologous domain evolution in which binding sites are obliterated. The C-terminal barrel incorporates the zinc ion that binds and activates Hcy. The zinc-binding site in MetE is distinguished from the (Cys)(3)Zn site in the related enzymes, MetH and betaine-homocysteine methyltransferase, by its position in the barrel and by the metal ligands, which are histidine, cysteine, glutamate, and cysteine in the resting form of MetE. Hcy associates at the face of the metal opposite glutamate, which moves away from the zinc in the binary E.Hcy complex. The folate substrate is not intimately associated with the N-terminal barrel; instead, elements from both barrels contribute binding determinants in a binary complex in which the folate substrate is incorrectly oriented for methyl transfer. Atypical locations of the Hcy and folate sites in the C-terminal barrel presumably permit direct interaction of the substrates in a ternary complex. Structures of the binary substrate complexes imply that rearrangement of folate, perhaps accompanied by domain rearrangement, must occur before formation of a ternary complex that is competent for methyl transfer.


Subject(s)
Evolution, Molecular , Methyltransferases/genetics , Methyltransferases/metabolism , Thermotoga maritima/genetics , Amino Acid Sequence , Escherichia coli/enzymology , Escherichia coli/genetics , Gene Duplication , Molecular Sequence Data , Peptide Fragments/metabolism , Substrate Specificity , Thermotoga maritima/enzymology
6.
Biochemistry ; 43(18): 5341-51, 2004 May 11.
Article in English | MEDLINE | ID: mdl-15122900

ABSTRACT

Betaine-homocysteine S-methyltransferase (BHMT) is a zinc-dependent enzyme that catalyzes the transfer of a methyl group from glycine betaine (Bet) to homocysteine (Hcy) to form dimethylglycine (DMG) and methionine (Met). Previous studies in other laboratories have indicated that catalysis proceeds through the formation of a ternary complex, with a transition state mimicked by the inhibitor S-(delta-carboxybutyl)-l-homocysteine (CBHcy). Using changes in intrinsic tryptophan fluorescence to determine the affinity of human BHMT for substrates, products, or CBHcy, we now demonstrate that the enzyme-substrate complex reaches its transition state through an ordered bi-bi mechanism in which Hcy is the first substrate to bind and Met is the last product released. Hcy, Met, and CBHcy bind to the enzyme to form binary complexes with K(d) values of 7.9, 6.9, and 0.28 microM, respectively. Binary complexes with Bet and DMG cannot be detected with fluorescence as a probe, but Bet and DMG bind tightly to BHMT-Hcy to form ternary complexes with K(d) values of 1.1 and 0.73 microM, respectively. Mutation of each of the seven tryptophan residues in human BHMT provides evidence that the enzyme undergoes two distinct conformational changes that are reflected in the fluorescence of the enzyme. The first is induced when Hcy binds, and the second, when Bet binds. As predicted by the crystal structure of BHMT, the amino acids Trp44 and Tyr160 are involved in binding Bet, and Glu159 in binding Hcy. Replacing these residues by site-directed mutagenesis significantly reduces the catalytic efficiency (V(max)/K(m)) of the enzyme. Replacing Tyr77 with Phe abolishes enzyme activity.


Subject(s)
Homocysteine/analogs & derivatives , Methyltransferases/chemistry , Methyltransferases/genetics , Mutagenesis, Site-Directed , Sarcosine/analogs & derivatives , Tryptophan/chemistry , Betaine/metabolism , Betaine-Homocysteine S-Methyltransferase , Binding Sites/genetics , Catalysis , Enzyme Inhibitors/chemistry , Glutamic Acid/genetics , Homocysteine/chemistry , Homocysteine/metabolism , Humans , Kinetics , Ligands , Methionine/metabolism , Methyltransferases/antagonists & inhibitors , Protein Binding/genetics , Sarcosine/chemistry , Sarcosine/metabolism , Spectrometry, Fluorescence/methods , Substrate Specificity/genetics , Tryptophan/genetics , Tyrosine/genetics
7.
Proc Natl Acad Sci U S A ; 101(11): 3729-36, 2004 Mar 16.
Article in English | MEDLINE | ID: mdl-14752199

ABSTRACT

B(12)-dependent methionine synthase (MetH) is a large modular enzyme that utilizes the cobalamin cofactor as a methyl donor or acceptor in three separate reactions. Each methyl transfer occurs at a different substrate-binding domain and requires a different arrangement of modules. In the catalytic cycle, the cobalamin-binding domain carries methylcobalamin to the homocysteine (Hcy) domain to form methionine and returns cob(I)alamin to the folate (Fol) domain for remethylation by methyltetrahydrofolate (CH(3)-H(4)folate). Here, we describe crystal structures of a fragment of MetH from Thermotoga maritima comprising the domains that bind Hcy and CH(3)-H(4)folate. These substrate-binding domains are (beta alpha)(8) barrels packed tightly against one another with their barrel axes perpendicular. The properties of the domain interface suggest that the two barrels remain associated during catalysis. The Hcy and CH(3)-H(4)folate substrates are bound at the C termini of their respective barrels in orientations that position them for reaction with cobalamin, but the two active sites are separated by approximately 50 A. To complete the catalytic cycle, the cobalamin-binding domain must travel back and forth between these distant active sites.


Subject(s)
5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/chemistry , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/metabolism , Amino Acid Sequence , Binding Sites , Molecular Sequence Data , Protein Structure, Tertiary , Thermotoga maritima/chemistry , Thermotoga maritima/enzymology , Thermotoga maritima/metabolism , Vitamin B 12/metabolism
8.
J Biol Chem ; 278(51): 51863-71, 2003 Dec 19.
Article in English | MEDLINE | ID: mdl-14506262

ABSTRACT

The bacterial enzyme, glycerol-3-phosphate cytidylyltransferase (GCT), is a model for mammalian cytidylyltransferases and is a member of a large superfamily of nucleotidyltransferases. Dimeric GCT from Bacillus subtilis displays unusual negative cooperativity in substrate binding and appears to form products only when both active sites are occupied by substrates. Here we describe a complex of GCT with the product, CDP-glycerol, in a crystal structure in which bound sulfate serves as a partial mimic of the second product, pyrophosphate. Binding of sulfate to form a pseudo-ternary complex is observed in three of the four chains constituting the asymmetric unit and is accompanied by a backbone rearrangement at Asp11 and ordering of the C-terminal helix. Comparison with the CTP complex of GCT, determined previously, reveals that in the product complex the active site closes around the glycerol phosphate moiety with a concerted motion of the segment 37-47 that includes helix B. This rearrangement allows lysines 44 and 46 to interact with the glycerol and cytosine phosphates of CDP-glycerol. Binding of CDP-glycerol also induces smaller movements of residues 92-100. Roles of lysines 44 and 46 in catalysis have been confirmed by mutagenesis of these residues to alanine, which decreases Vmax(app) and has profound effects on the Km(app) for glycerol-3-phosphate.


Subject(s)
Bacterial Proteins/chemistry , Nucleotidyltransferases/chemistry , Allosteric Regulation , Amino Acid Substitution , Bacillus subtilis/enzymology , Catalysis , Kinetics , Nucleoside Diphosphate Sugars/chemistry , Protein Binding , Protein Structure, Secondary , Sulfates/chemistry
9.
Proc Natl Acad Sci U S A ; 100(14): 8156-63, 2003 Jul 08.
Article in English | MEDLINE | ID: mdl-12832615

ABSTRACT

In the course of catalysis or signaling, large multimodular proteins often undergo conformational changes that reposition the modules with respect to one another. The mechanisms that direct the reorganization of modules in these proteins are of considerable importance, but distinguishing alternate conformations is a challenge. Cobalamin-dependent methionine synthase (MetH) is a 136-kDa multimodular enzyme with a cobalamin chromophore; the color of the cobalamin reflects the conformation of the protein. The enzyme contains four modules and catalyzes three different methyl transfer reactions that require different arrangements of these modules. Two of these methyl transfer reactions occur during turnover, when homocysteine is converted to methionine by using a methyl group derived from methyltetrahydrofolate. The third reaction is occasionally required for reactivation of the enzyme and uses S-adenosyl-L-methionine as the methyl donor. The absorbance properties of the cobalamin cofactor have been exploited to assign conformations of the protein and to probe the effect of ligands and mutations on the distribution of conformers. The results imply that the methylcobalamin form of MetH exists as an ensemble of interconverting conformational states. Differential binding of substrates or products alters the distribution of conformers. Furthermore, steric conflicts disfavor conformers that juxtapose a methyl group on substrate with one on methylcobalamin. These results suggest that the methylation state of the cobalamin will influence the distribution of conformers during turnover.


Subject(s)
5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Vitamin B 12/analogs & derivatives , Vitamin B 12/chemistry , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/genetics , Amino Acid Substitution , Catalysis , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Homocysteine/metabolism , Methionine/biosynthesis , Methylation , Mutation, Missense , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , S-Adenosylmethionine/metabolism , Spectrophotometry, Ultraviolet , Structure-Activity Relationship , Substrate Specificity , Temperature , Tetrahydrofolates/metabolism
10.
Structure ; 10(9): 1159-71, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12220488

ABSTRACT

Betaine-homocysteine methyl transferase (BHMT) catalyzes the synthesis of methionine from betaine and homocysteine (Hcy), utilizing a zinc ion to activate Hcy. BHMT is a key liver enzyme that is important for homocysteine homeostasis. X-ray structures of human BHMT in its oxidized (Zn-free) and reduced (Zn-replete) forms, the latter in complex with the bisubstrate analog, S(delta-carboxybutyl)-L-homocysteine, were determined at resolutions of 2.15 A and 2.05 A. BHMT is a (beta/alpha)(8) barrel that is distorted to construct the substrate and metal binding sites. The zinc binding sequences G-V/L-N-C and G-G-C-C are at the C termini of strands beta6 and beta8. Oxidation to the Cys217-Cys299 disulfide and expulsion of Zn are accompanied by local rearrangements. The structures identify Hcy binding fingerprints and provide a prototype for the homocysteine S-methyltransferase family.


Subject(s)
Methyltransferases/chemistry , Methyltransferases/metabolism , Zinc/metabolism , Amino Acid Sequence , Betaine-Homocysteine S-Methyltransferase , Binding Sites , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Conformation , Sequence Homology, Amino Acid , Static Electricity , Substrate Specificity
12.
Nat Struct Biol ; 9(1): 53-6, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11731805

ABSTRACT

B(12)-dependent methionine synthase (MetH) from Escherichia coli is a large modular protein that uses bound cobalamin as an intermediate methyl carrier. Major domain rearrangements have been postulated to explain how cobalamin reacts with three different substrates: homocysteine, methyltetrahydrofolate and S-adenosylmethionine (AdoMet). Here we describe the 3.0 A structure of a 65 kDa C-terminal fragment of MetH that spans the cobalamin- and AdoMet-binding domains, arranged in a conformation suitable for the methyl transfer from AdoMet to cobalamin that occurs during activation. In the conversion to the activation conformation, a helical domain that capped the cofactor moves 26 A and rotates by 63 degrees, allowing formation of a new interface between cobalamin and the AdoMet-binding (activation) domain. Interactions with the MetH activation domain drive the cobalamin away from its binding domain in a way that requires dissociation of the axial cobalt ligand and, thereby, provide a mechanism for control of the distribution of enzyme conformations.


Subject(s)
5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/chemistry , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/metabolism , Escherichia coli/enzymology , Binding Sites , Crystallography, X-Ray , Enzyme Activation , Models, Molecular , Movement , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , S-Adenosylmethionine/metabolism , Vitamin B 12/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...